Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
2.
Neurotoxicology ; 28(6): 1092-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17686523

RESUMO

Paclitaxel-induced sensory neuropathy is a problematic side-effect of cancer chemotherapy. Previous studies in rodents have shown paclitaxel treatment to have many effects on different parts of the peripheral nervous system, but those responsible for its bothersome clinical side-effects are still unclear. In the current study, we sought to obtain information about the involvement of sensory neurons in paclitaxel neurotoxicity at the level of the dorsal root ganglion. Rats were treated with a clinically relevant dose of paclitaxel (87.5mg/m(2) weekly for a total of nine doses) to induce a sensory neuropathy; then their L5 dorsal root ganglia were studied by morphometry and immunohistochemistry. Paclitaxel treatment was generally well tolerated, and slowed conduction velocity and prolonged conduction latencies in the peripheral sensory nerves without altering conduction in the central or motor pathways of the H-reflex arc. In the L5 dorsal root ganglion, nucleolus size and the number of neurons with eccentric nuclei were increased only in a subpopulation of dorsal root ganglion neurons with cell body cross-sectional areas greater than 1750 microm(2), which made up less than 10% of the total population. Paclitaxel treatment increased immunohistochemical staining for activating transcription factor-3 (ATF-3), c-Jun and neuropeptide Y (NPY) but only in a small percentage of neuronal cell bodies and mainly in those with large cell bodies. In conclusion, we have demonstrated that nucleolar enlargement, nuclear eccentricity, ATF-3, c-Jun and NPY are neuronal markers of paclitaxel-induced sensory neuropathy, however, these axotomy-like cell body reactions are infrequent and occur in mainly large-sized sensory neurons.


Assuntos
Nucléolo Celular/patologia , Gânglios Espinais/patologia , Imuno-Histoquímica , Neurônios Aferentes/patologia , Síndromes Neurotóxicas/patologia , Doenças do Sistema Nervoso Periférico/patologia , Fator 3 Ativador da Transcrição/análise , Animais , Antineoplásicos Fitogênicos , Tamanho Celular , Modelos Animais de Doenças , Feminino , Gânglios Espinais/química , Gânglios Espinais/fisiopatologia , Reflexo H , Condução Nervosa , Neurônios Aferentes/química , Neuropeptídeo Y/análise , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/fisiopatologia , Paclitaxel , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/fisiopatologia , Proteínas Proto-Oncogênicas c-jun/análise , Ratos , Ratos Wistar , Tempo de Reação , Regulação para Cima
3.
Cancer Res ; 54(3): 629-31, 1994 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8306321

RESUMO

This study compared the effect on sensory nerve conduction velocity in the hind limb of chronically treated age-matched rats of a novel lipophilic p.o. platinum complex [bisacetatoamminedichlorocyclohexylamine-platinum(IV)], with that of neurotoxic platinum complexes cisplatin and tetraplatin. Tetraplatin (i.p.) first caused slowing of sensory nerve conduction (-14.4% of controls; P < 0.05) at a dose (1 mg/kg) that was free of constitutional toxicity. Cisplatin (i.p.) first caused slowing of sensory nerve conduction (-17.5% of control; P < 0.05) at a dose (2 mg/kg) approximating the maximal tolerated dose. Bisacetatoamminedichlorocyclohexylamineplatinum(IV) (p.o.) showed no slowing of sensory nerve conduction either after 20.5 weeks of treatment at a dose (25 mg/kg) approximating the maximal tolerated dose or after 6 weeks of treatment at a dose (50 mg/kg) which eventually proved intolerable. In conclusion, p.o. bisacetatoamminedichlorocyclohexylamineplatinum(IV) shows a lack of neurotoxicity compared to parenterally administered cisplatin and tetraplatin at the maximal tolerated dose in rats.


Assuntos
Antineoplásicos/toxicidade , Cisplatino/toxicidade , Doenças do Sistema Nervoso/induzido quimicamente , Compostos Organoplatínicos/toxicidade , Administração Oral , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Injeções Intraperitoneais , Doenças do Sistema Nervoso/fisiopatologia , Condução Nervosa/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/fisiologia , Ratos , Ratos Wistar
4.
Cancer Res ; 54(15): 4118-22, 1994 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-8033145

RESUMO

JM216 is a novel antitumor platinum(IV) complex displaying oral activity, dose-limiting myelosuppression, and a lack of nephro- and neurotoxicity in rodents. It has been selected for clinical evaluation. The schedule dependency of its antitumor action against a murine (ADJ/PC6 plasmacytoma) and a human tumor model (PXN109T/C ovarian carcinoma xenograft) was studied in vivo. Single dose (q21d), once a day dosing for 5 consecutive days (q21d or q28d), and once a day dosing indefinitely (chronic daily dosing) administration schedules were compared. Against the murine ADJ/PC6 plasmacytoma, daily x5 administration improved the tolerance, antitumor potency, and therapeutic index of oral JM216, compared to single dose administration, whereas no advantage was found for fractionating cisplatin dosages. Against the PXN109T/C human ovarian carcinoma xenograft, oral JM216, given at dose levels delivering a equivalent total dose on single dose (200 mg/kg q21d), daily x5 (40 mg/kg/day q21d) and chronic daily dosing (9.5 mg/kg/d) schedules, showed superior tumor growth delays (55 +/- 15 days; P < 0.05) and maximal tumor regression (10 +/- 11% of initial tumor volume; P < 0.001) with the daily x5 schedule. Gastrointestinal toxicity (P < 0.05) and mild nephrotoxicity (P < 0.01) complicated the chronic daily dosing schedule, while leukopenia (P < 0.02) and thrombocytopenia (P < 0.01) were dose-limiting for the single dose and daily x5 administration, respectively. Peak plasma ultrafiltrate (PUF) platinum levels were below cytotoxic levels (PUF Cmax, 0.11 +/- 0.066 mg/l) at the maximally tolerable dose for the chronic dosing schedule (9.5 mg/kg). Peak PUF platinum levels did not increase significantly with a 5-fold increase in dosage from 40 mg/kg (PUF Cmax 1.5 +/- 0.11 mg/l) to 200 mg/kg (PUF Cmax, 2.4 +/- 0.44 mg/l; P > 0.05). In conclusion, these data demonstrate antitumor schedule dependency for oral JM216 in vivo, independently in two tumor model systems, and with nonlinear pharmacokinetics after its oral administration to mice. Optimal antitumor activity, tolerance, and pharmacokinetics occurred with daily x5 dosing, and this has prompted the clinical evaluation of this administration schedule.


Assuntos
Adenocarcinoma Papilar/tratamento farmacológico , Antineoplásicos/administração & dosagem , Compostos Organoplatínicos/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Adenocarcinoma Papilar/metabolismo , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Peso Corporal/efeitos dos fármacos , Cisplatino/administração & dosagem , Cisplatino/farmacocinética , Esquema de Medicação , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Leucopenia/induzido quimicamente , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Compostos Organoplatínicos/efeitos adversos , Compostos Organoplatínicos/farmacocinética , Neoplasias Ovarianas/metabolismo , Trombocitopenia/induzido quimicamente , Transplante Heterólogo , Células Tumorais Cultivadas
5.
Cancer Res ; 53(11): 2581-6, 1993 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-8388318

RESUMO

The cytotoxicity of a novel platinum(IV) complex, bis-acetato-amminedichloro-cyclohexylamine platinum(IV) (JM216), has been evaluated in vitro against a panel of human tumor cell lines (predominantly ovarian) representative of models of intrinsic and acquired to cisplatin. In addition, the activity of JM216 administered by the p.o. route has been determined in vivo using the murine ADJ/PC6 plasmacytoma and four human ovarian carcinoma xenograft lines. In vitro, against seven human ovarian carcinoma cell lines, JM216 showed similar cytotoxicity and pattern of cytotoxicity to cisplatin (mean 50% inhibitory concentrations of 3.5 microM for cisplatin and 1.7 microM for JM216). The cytotoxicity of JM216 was more dependent on the time of drug exposure than that of cisplatin, suggesting that extended split-dosing rather than a single bolus administration might be a more appropriate schedule in patients. Using six pairs of acquired cisplatin-resistant and parent human tumor cell lines (four ovarian, one testicular, and one cervical) JM216 exhibited non-cross-resistance (resistance factor of < 1.5) in three whereas tetraplatin exhibited partial or full cross-resistance in all six pairs. Notably, in two of the acquired cisplatin-resistant lines (41McisR and HX/155cisR) where JM216 retained activity, resistance has previously shown to be due primarily to reduced platinum uptake. In vivo, following p.o. administration using the cisplatin-sensitive murine ADJ/PC6 plasmacytoma, JM216 showed antitumor selectivity far superior to that observed for either cisplatin, carboplatin, or tetraplatin. Across four human ovarian carcinoma xenografts of widely differing sensitivity to cisplatin and carboplatin, JM216 exhibited p.o. activity, broadly comparable to that observed for i.v. administered cisplatin and carboplatin and markedly superior to i.p. administered tetraplatin. These antitumor properties suggest that JM216 provides a structural lead to platinum complexes which may circumvent transport-determined acquired resistance to cisplatin and is a suitable candidate as an p.o. administrable platinum complex for phase I clinical trial.


Assuntos
Neoplasias/tratamento farmacológico , Compostos Organoplatínicos/farmacologia , Animais , Carboplatina/farmacologia , Cisplatino/farmacologia , Resistência a Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Leucemia L1210/tratamento farmacológico , Masculino , Camundongos , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Compostos Organoplatínicos/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Plasmocitoma/tratamento farmacológico , Neoplasias Testiculares/tratamento farmacológico , Fatores de Tempo , Transplante Heterólogo , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/tratamento farmacológico
6.
J Clin Oncol ; 8(5): 779-83, 1990 May.
Artigo em Inglês | MEDLINE | ID: mdl-1692088

RESUMO

In a retrospective analysis, serial lung function tests from 81 patients receiving bleomycin were studied to determine the accuracy of carbon monoxide diffusing capacity (DLCO) as a predictor of clinically significant bleomycin lung. Six of 81 patients developed clinically significant bleomycin lung, and the DLCO predicted its development in only one patient (sensitivity, one of six patients; 16.7%). Respiratory symptoms and chest x-ray abnormalities were the earliest manifestations in the other five patients. Seventy-five of 81 patients did not develop clinically significant bleomycin lung, and 12 of these had major falls (greater than or equal to 35% pretreatment level) in DLCO (specificity, 63 of 75 patients; 84.0%). In eight patients, bleomycin was continued after a major fall in DLCO, and none developed clinically significant lung toxicity. In this study, the DLCO failed to predict the development of serious bleomycin lung toxicity in all but one case. Furthermore, in some patients, it would appear that bleomycin may be stopped inappropriately after low DLCO measurements. It is important to monitor for respiratory symptoms and chest x-ray abnormalities during bleomycin treatment as these will be the earliest signs of lung toxicity in most cases.


Assuntos
Bleomicina/efeitos adversos , Monóxido de Carbono , Pulmão/efeitos dos fármacos , Capacidade de Difusão Pulmonar/efeitos dos fármacos , Adolescente , Adulto , Idoso , Bleomicina/administração & dosagem , Reações Falso-Positivas , Feminino , Humanos , Pulmão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Radiografia Torácica , Estudos Retrospectivos
7.
J Clin Oncol ; 15(7): 2691-700, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9215842

RESUMO

PURPOSE: We aimed to determine the maximum-tolerated dose (MTD) clinical toxicities, pharmacokinetics, and pharmacodynamics of oral JM216 given once daily for 5 days to cancer patients. PATIENTS AND METHODS: Patients who fulfilled standard phase I trial criteria were enrolled. Oral JM216 was given at doses based on patient body-surface area, on an empty stomach, once daily for 5 consecutive days, as 10-, 50-, and 200-mg hard gelatin capsules and with oral antiemetics. The pharmacokinetics of platinum were studied on days 1 and 5 of the first treatment course using atomic absorption spectrophotometry (AAS). RESULTS: Thirty-two patients received 94 courses of oral JM216 at doses that ranged from 30 to 140 mg/m2 body-surface area for 5 consecutive days. The MTD was 140 mg/m2/d. The dose-limiting toxicities were thrombocytopenia and neutropenia. Hematotoxicity was reversible (nadir, 17 to 21 days; recovery, 28 days), noncumulative, and dependent on the dose and history of previous therapy. There were two instances of neutropenic sepsis. Two-thirds of patients experienced mild nausea, vomiting, or diarrhea. There was no ototoxicity, neurotoxicity, nephrotoxicity, or objective tumor responses. There was a significant correlation between JM216 dose and the day 1 and 5 plasma ultrafiltrate area under the concentration-time curve (AUC; r = .78), which indicates linear pharmacokinetics. There was considerable intersubject pharmacokinetic and pharmacodynamic variability, but a significant sigmoidal relationship between the plasma ultrafiltrate AUC and severity of thrombocytopenia (R2 = .83). CONCLUSION: We recommend JM216 doses of 100 and 120 mg/m2/d x 5 for previously treated and untreated patients, respectively, for phase II trials.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Administração Oral , Adulto , Idoso , Antineoplásicos/efeitos adversos , Área Sob a Curva , Esquema de Medicação , Feminino , Gastroenteropatias/induzido quimicamente , Doenças Hematológicas/induzido quimicamente , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Compostos Organoplatínicos , Índice de Gravidade de Doença , Ultrafiltração
8.
Cancer Chemother Pharmacol ; 56(4): 391-9, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15887017

RESUMO

Peripheral neuropathy is induced by multiple doses of oxaliplatin and interferes with the clinical utility of the drug in patients with colorectal cancer. In this study, we sought to determine whether cell loss or selective neuronal damage was the basis for the peripheral neuropathy caused by oxaliplatin. Adult female rats were given 1.85 mg/kg oxaliplatin twice per week for 8 weeks. Nerve conduction and L5 dorsal root ganglia (DRG) were studied 1 week after the completion of all treatment. No mortality occurred during oxaliplatin treatment, but the rate of body weight gain was reduced compared to age-matched vehicle-treated controls. Oxaliplatin slowed conduction velocity and delayed conduction times in peripheral sensory nerves, without affecting central or motor nerve conduction. In L5 DRG, total numbers of neurons were unchanged by oxaliplatin, but there were significant reductions in neuronal size distribution, ganglion volume, average cell size and the relative frequency of large cells. In addition, the relative frequency of small DRG cells was increased by oxaliplatin. Oxaliplatin significantly altered the size distribution and average cell body area of the predominantly large parvalbumin-immunoreactive DRG neurons without affecting the frequency of parvalbumin staining. On the contrary, neither the staining frequency nor the size distribution of the predominantly small substance P-immunoreactive DRG neurons was changed by oxaliplatin. In conclusion, oxaliplatin causes selective atrophy of a subpopulation of DRG neurons with predominantly large parvalbumin-expressing cells without inducing neuronal loss. Because DRG cell body size and axonal conduction velocity are positively correlated, neuronal atrophy may be the morphological basis for the development of decreased sensory nerve conduction velocity that characterizes oxaliplatin-induced peripheral neuropathy.


Assuntos
Antineoplásicos/toxicidade , Gânglios Espinais/efeitos dos fármacos , Condução Nervosa/efeitos dos fármacos , Compostos Organoplatínicos/toxicidade , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Animais , Feminino , Oxaliplatina , Parvalbuminas/isolamento & purificação , Doenças do Sistema Nervoso Periférico/patologia , Ratos , Ratos Wistar , Substância P/isolamento & purificação
9.
Semin Oncol ; 26(3 Suppl 11): 13-8, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10458205

RESUMO

Cisplatin-based chemotherapy is effective in non-small cell lung cancer (NSCLC), although it prolongs survival only modestly. Single-agent docetaxel (Taxotere; Rhône-Poulenc Rorer, Antony, France) is highly active against NSCLC. The activity and tolerability of two docetaxel/ cisplatin regimens were therefore investigated in two multicenter phase II studies, one in Australia and one in France. Chemotherapy-naive patients with inoperable NSCLC received either docetaxel 75 mg/m2 on day 1 plus cisplatin 75 mg/m2 3 weekly (n = 47; Australian study) or docetaxel 75 mg/m2 on day 1 plus cisplatin 100 mg/m2 every 3 weeks for three cycles then every 6 weeks (n = 51; French study). The majority of the population (74%) had metastatic disease. Seventy-eight patients were evaluable for efficacy. Overall response rates were 36% (95% confidence interval, 25 to 47) in all evaluable patients and 34% in patients with metastases. Median duration of response was 6 months, with a 4-month median time to progression. Median survival time was 9 months, with a 1-year survival rate of 34%. A median of four (range, one to nine) treatment cycles were administered. Febrile neutropenia occurred in 14% of patients. Severe infection, which occurred in less than 7% of patients, led to two toxic deaths. Other severe toxicities were rare, with severe stomatitis and severe neurosensory side effects reported in 2% and 1%, respectively, of treated patients. No severe fluid retention occurred. Docetaxel/cisplatin, administered as two different schedules, is well tolerated and exhibits efficacy in the range of the most established combinations in the treatment of advanced NSCLC.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Paclitaxel/análogos & derivados , Taxoides , Adulto , Idoso , Carcinoma Pulmonar de Células não Pequenas/secundário , Cisplatino/administração & dosagem , Docetaxel , Esquema de Medicação , Feminino , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Paclitaxel/administração & dosagem , Análise de Sobrevida
10.
J Med Chem ; 40(7): 1090-8, 1997 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-9089330

RESUMO

A pair of enantiomeric Pt(II) complexes, [Pt(R-ahaz)Cl2] and [Pt(S-ahaz)Cl2] (ahaz = 3-aminohexahydroazepine), has been investigated for their ability to bind enantioselectively to DNA. Improved synthetic procedures were developed for preparing both the ligands and the Pt complexes. The structure of the complex of the S enantiomer was determined by X-ray crystallographic methods. Crystals of [Pt(S-ahaz)Cl2] are orthorhombic, space group P2(1)2(1)2(1), with a = 6.917(1) A, b = 11.167(1) A, c = 12.373(2) A, Z = 4, and the structure was refined to R = 0.023 (1505F). Molecular modeling techniques were used to investigate the role of steric interactions between the ligand and DNA in influencing the bifunctional binding of the two enantiomers, and it was found that the S enantiomer should bind more readily. The binding of the S enantiomer, to calf thymus DNA, was indeed found to be slightly greater than that for the R enantiomer though slightly less than that for cis-DDP. Assays of the proportion of monofunctional adducts showed that a substantially greater proportion of monofunctional adducts remained for the R enantiomer and cisplatin than for the S enantiomer. Each of the enantiomers was subjected to in vitro cytotoxicity assays using cultures of human bladder (BL13/0), lung and resistant lung (PC9 and PC9cisR), and prostate (DU145) cancer cells. The R enantiomer was found to be slightly more cytotoxic in the bladder cell line and may be less cytotoxic in the lung cell line but there were no significant differences in the resistant cell line nor in the prostate cell line. The two enantiomers were taken up equally by the bladder cancer cells.


Assuntos
Antineoplásicos/síntese química , DNA/metabolismo , Compostos Organometálicos/síntese química , Antineoplásicos/química , Antineoplásicos/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Compostos Organometálicos/química , Compostos Organometálicos/metabolismo , Estereoisomerismo , Células Tumorais Cultivadas
11.
J Med Chem ; 40(22): 3508-15, 1997 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-9357517

RESUMO

A series of chiral diaminedichloroplatinum(II) complexes derived from [Pt(ahaz)Cl2] (ahaz = 3-aminohexahydroazepine) have been synthesized and tested for cytotoxic activity. Novel synthetic pathways were developed to produce the structural derivatives of the ahaz ligand, with alkyl substituents on the exocyclic nitrogen atom. The platinum(II) complexes of these ligands were synthesized and characterized by NMR and CD spectroscopy, confirming isomeric and enantiomeric purity. The crystal structures of three of these complexes, [Pt(S-meahaz)-Cl2], [Pt(R-etahaz)Cl2], and [Pt(S-dimeahaz)Cl2] (meahaz = N-methylahaz, etahaz = N-ethylahaz, dimeahaz = N,N-dimethylahaz), have been determined to establish the orientation of the protons and alkyl substituents on the nitrogen donor atoms. In vitro assays of the cytotoxic activity of the complexes have revealed a significant and reproducible enantioselective trend with the R-enantiomers more active than the S-enantiomers in all cell lines. Increasing the steric bulk on the amine groups was found to have only a modest effect on activity. No enantioselectivity was observed in the binding of R- and S-[Pt(etahaz)Cl2] to calf-thymus DNA.


Assuntos
Antineoplásicos/farmacologia , DNA/efeitos dos fármacos , Compostos Organoplatínicos/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Bovinos , Dicroísmo Circular , Cristalografia por Raios X , DNA/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Compostos Organoplatínicos/química , Compostos Organoplatínicos/metabolismo , Estereoisomerismo , Células Tumorais Cultivadas
12.
Expert Opin Investig Drugs ; 10(1): 119-28, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11116285

RESUMO

Lobaplatin (D-19466) is a diastereometric mixture of platinum(II) complexes containing a 1,2-bis(aminomethyl)cyclobutane stable ligand and lactic acid as the leaving group. Its antitumour activity results from the formation of DNA-drug adducts, mainly as GG and AG intra-strand cross-links. Lobaplatin influences the expression of the c-myc gene, which is involved in oncogenesis, apoptosis and cell proliferation. Lobaplatin has activity in a wide range of preclinical tumour models and appears to overcome tumour resistance to cisplatin and carboplatin in some of these models. In the body, lobaplatin remains largely intact until removed by glomerular filtration. Exposure of the body to lobaplatin (AUC) correlates with dose, creatinine clearance and the degree of thrombocytopoenia. Phase I clinical trials of three quite different administration schedules found the same dose-limiting toxicity (thrombocytopoenia) and similar maximum tolerated doses (60 mg/m(2) per 3 - 4 weeks). In Phase II trials, lobaplatin showed activity in patients with a variety of tumour types. Many of the patients who responded to lobaplatin may also have responded to cisplatin and carboplatin because they had had no prior chemotherapy or had a prolonged remission after earlier treatment. In conclusion, lobaplatin is a new platinum drug, which overcomes some forms of cisplatin resistance in preclinical tumour models. Several potential clinical applications remain unexplored, such as its use in relapsed testicular cancer and in combination with other cancer chemotherapeutic agents and ionising radiation.


Assuntos
Antineoplásicos/uso terapêutico , Ciclobutanos/uso terapêutico , Compostos Organoplatínicos/uso terapêutico , Animais , Ensaios Clínicos como Assunto , Ciclobutanos/farmacocinética , Ciclobutanos/farmacologia , Humanos , Compostos Organoplatínicos/farmacocinética , Compostos Organoplatínicos/farmacologia
13.
Drug Saf ; 13(4): 228-44, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8573296

RESUMO

Since the discovery of the biologically active platinum complexes 30 years ago, 2 agents have become widely established in clinical oncology practice. Both cisplatin and carboplatin are platinum(II) complexes with 2 ammonia groups in the cis- position. However, they differ in their solubility, chemical reactivity, dichloride or alicyclic oxygenated leaving groups, pharmacokinetics and toxicology. Cisplatin causes severe renal tubular damage and reduces glomerular filtration, and requires concurrent saline hydration and mannitol diuresis to eliminate potentially lethal and unacceptable damage to the kidneys. Carboplatin, at conventional doses, causes no decrease in glomerular filtration and only minor transient elevations in urinary enzymes. Cisplatin is the most emetic cancer drug in common use, while nausea and vomiting associated with carboplatin are moderately severe. Serotonin release from enterochromaffin gut mucosal cells and stimulation of serotonin 5-HT3-receptors mediates acute emesis. Selective inhibitors of the 5-HT3-receptor protect against cisplatin- and carboplatin-induced nausea and vomiting. Peripheral neurotoxicity is the most dose-limiting problem associated with cisplatin. Loss of vibration sense, paraesthesia and sensory ataxia comes on after several treatment cycles. Carboplatin, however, is relatively free from peripheral neurotoxicity. Audiometry shows cisplatin-induced ototoxicity in 75 to 100% of patients, which may be associated with tinnitus and hearing loss. Ototoxicity is rare with conventional dose carboplatin therapy. Monitoring hearing with audiograms may identify early signs before significant impairment occurs. Cisplatin causes mild haematological toxicity to all 3 blood lineages. Haematological toxicity is dose-limiting for carboplatin, with thrombocytopenia being a greater problem than leucopenia. Although carboplatin is not toxic to the kidney, renal function markedly affects the severity of carboplatin-induced thrombocytopenia. The major clearance mechanism of cisplatin is irreversible binding in plasma and tissues, while carboplatin is cleared by glomerular filtration. Metabolism of cisplatin to aqua, amino acid and protein species is extensive, whereas carboplatin exists mainly as the free unchanged form. Strong relationships between carboplatin renal clearance, glomerular filtration rate, area under the plasma concentration-time curve (AUC) of filterable platinum and severity of thrombocytopenia have prompted dose adjustment according to renal function. New analogues such as JM216 offer the potential advantages of oral administration and few nonhaematological toxicities. Analogues based on the diaminocyclohexane ligand have encountered problematic neurotoxicity.


Assuntos
Antineoplásicos/efeitos adversos , Carboplatina/efeitos adversos , Cisplatino/efeitos adversos , Antineoplásicos/sangue , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Carboplatina/sangue , Carboplatina/farmacocinética , Carboplatina/uso terapêutico , Cisplatino/sangue , Cisplatino/farmacocinética , Cisplatino/uso terapêutico , Cóclea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Taxa de Filtração Glomerular/efeitos dos fármacos , Perda Auditiva Neurossensorial/induzido quimicamente , Doenças Hematológicas/induzido quimicamente , Humanos , Túbulos Renais/efeitos dos fármacos , Náusea/induzido quimicamente , Sistema Nervoso Periférico/efeitos dos fármacos , Receptores de Serotonina/efeitos dos fármacos , Relação Estrutura-Atividade , Vômito/induzido quimicamente
14.
Cancer Chemother Pharmacol ; 28(6): 409-13, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1934244

RESUMO

Although the antitumour agent flavone-8-acetic acid (FAA) exhibits remarkable activity against murine solid tumours, its clinical use has a number of pharmacological drawbacks, including low dose potency and dose-dependent pharmacokinetics. Xanthenone-4-acetic acid (XAA) and its 5,6-dimethyl derivative (5,6-MeXAA) were synthesised during a search for better analogues of FAA. The maximal tolerated doses (MTDs) of 5,6-MeXAA, XAA and FAA in BDF1 mice were 99, 1,090 and 1,300 mumol/kg, respectively. At the MTD, 5,6-MeXAA displayed the following pharmacokinetic properties: maximal plasma concentration, 600 microM; mean residence time, 4.9 h; AUC, 2,400 mumol h 1-1; and volume of steady-state distribution, 0.2 l/kg. All compounds displayed nonlinear elimination kinetics at the MTD, but when the logarithm of the AUC was plotted against that of the delivered dose, the slope of the regression line for 5,6-MeXAA was found to be 1.2 as opposed to 1.4 for XAA and 1.98 for FAA. 5,6-MeXAA thus showed only a slight deviation from dose-independent kinetics. 5,6-MeXAA bound to plasma proteins in a manner similar to that exhibited by FAA, although the plasma concentration of free drug was lower for the former than for the latter. As a consequence, the calculated maximal free drug concentration for 5,6-MeXAA in plasma was 23 times lower than that for FAA.


Assuntos
Antineoplásicos/sangue , Flavonoides/sangue , Xantenos/sangue , Xantonas , Animais , Proteínas Sanguíneas/análise , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos , Ligação Proteica/efeitos dos fármacos
15.
Cancer Chemother Pharmacol ; 46(5): 343-50, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11127937

RESUMO

PURPOSE: The lipophilic cation [Au(I)(dppe)2]+ [where dppe is 1,2-bis(diphenylphosphino)ethane] has previously demonstrated potent in vitro antitumour activity. We wished to determine the physicochemical basis for the cellular uptake of this drug, as well as of analogues including the 1:2 adducts of Au(I) with 1,2-bis(di-n-pyridylphosphino)ethane (dnpype; n = 2, 3 and 4), and to compare in vitro and in vivo antitumour activity. METHODS AND RESULTS: Logarithmic IC50 values for the CH-1 cell line bore a parabolic dependence on drug lipophilicity, as measured either by high-performance liquid chromatography or by n-octanol-water partition. Cellular uptake of drug, as measured by inductively coupled plasma mass spectrometry, varied by over three orders of magnitude over the series. Logarithmic uptake had a parabolic dependence on drug lipophilicity but a linear relationship to logarithmic IC50 values. Free drug concentrations were determined under the culture conditions and logarithmic free drug IC50 values and uptake rates were linearly related to lipophilicity. Uptake of drug in vivo in tissue from murine colon 38 tumours was approximately proportional to the dose administered. Host toxicity varied according to lipophilicity with the most selective compound having an intermediate value. This compound was also the most active of those tested in vivo, giving a growth delay of 9 days following daily intraperitoneal dosing (10 days) at 4 micromol kg(-1) day(-1). It was also significantly more active than another lipophilic cation, MKT-077. CONCLUSIONS: Alteration of lipophilicity of aromatic cationic antitumour drugs greatly affects cellular uptake and binding to plasma proteins. Changes in lipophilicity also affect host toxicity, and optimal lipophilicity may be a critical factor in the design of analogues with high antitumour activity.


Assuntos
Antineoplásicos/farmacocinética , Compostos Organometálicos/farmacocinética , Compostos Organofosforados/farmacocinética , Neoplasias Ovarianas/metabolismo , Fosfinas/farmacocinética , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Divisão Celular/efeitos dos fármacos , Fenômenos Químicos , Físico-Química , Feminino , Humanos , Lipídeos/química , Camundongos , Transplante de Neoplasias , Compostos Organoáuricos , Compostos Organometálicos/química , Compostos Organometálicos/farmacologia , Compostos Organofosforados/química , Compostos Organofosforados/farmacologia , Fosfinas/química , Fosfinas/farmacologia , Ligação Proteica , Células Tumorais Cultivadas
16.
Cancer Chemother Pharmacol ; 28(6): 414-9, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1934245

RESUMO

Treatment of C57Bl/6 x DBA/2 mice with the maximal tolerated dose of flavone-8-acetic acid (FAA, 1300 mumol/kg), xanthenone-4-acetic acid (XAA, 1090 mumol/kg), or its dose-potent derivative 5,6-dimethyl-xanthenone-4-acetic acid (5,6-MeXAA, 100 mumol/kg) resulted within 24 h in a dramatic reduction in the number of circulating lymphocytes, an elevation in haemoglobin concentrations and a reduction in platelet numbers. Neutrophil counts either remained unchanged or were slightly elevated. All three compounds caused a marked loss of cells in the thymus. Examination of histological sections of thymus at 48 h following treatment with XAA revealed a selective depletion of cortical thymocytes and no effects on the epithelium or other thymic structures. A transient decrease in cell numbers was seen in the spleen and femoral bone marrow, with recovery to normal levels occurring within 3 days. The number of haemopoietic stem cells, colony-forming units in culture (CFU-c), in the femoral bone marrow increased after drug administration despite the occurrence of a decrease in the overall number of cells in the femur. In contrast to the increase in CFU-c numbers seen in vivo, 2 h exposure of bone-marrow cells to FAA, XAA or 5,6-MeXAA in vitro resulted in a decrease in the surviving fraction of CFU-c. The results are consistent with the hypothesis that the in vivo haematological effects of these compounds are indirect, perhaps being mediated through the induction of cytokines, and contrast with the haematological effects of conventional antitumour agents. The biochemical and haematological effects are unlikely to be the cause of the acute toxicity observed for these compounds.


Assuntos
Antineoplásicos/toxicidade , Células Sanguíneas/efeitos dos fármacos , Sangue/efeitos dos fármacos , Flavonoides/toxicidade , Xantenos/toxicidade , Xantonas , Animais , Medula Óssea/efeitos dos fármacos , Células da Medula Óssea , Ensaio de Unidades Formadoras de Colônias , Linfócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA
17.
Cancer Chemother Pharmacol ; 36(6): 451-8, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7554035

RESUMO

JM216 [bis-acetato-ammine-dichloro-cyclo-hexylamine-platinum (IV)] is an oral platinum complex with in vivo activity against murine and human tumor models and a lack of nephro- and neurotoxicity in rodents. During a phase I study of a single-dose schedule, JM216 was given in dry-filled hard gelatin capsules by mouth without hydration or diuresis. In all, 37 patients were given a total of 88 courses at doses ranging from 60 to 700 mg/m2. The study was stopped before the MTD was reached because of nonlinear pharmacokinetics. Myelosuppression was manifest by leucopenia or thrombocytopenia and showed marked variability at 420-700 mg/m2. Vomiting was mild and controllable by antiemetics in approximately 50% of courses. The onset of vomiting was delayed to 4 h after during ingestion. There was no nephro-, oto- or neuro-toxicity. A partial response was recorded in a patient with recurrent ovarian cancer, and significant falls in plasma tumour markers (CA125) were seen in two further cases. Plasma pharmacokinetics were linear and showed moderate interpatient variability at dose levels of < or = 120 mg/m2. At dose levels of > or = 200 mg/m2, Cmax and AUC increased less than proportionally to dose. This was associated with greater interpatient pharmacokinetic variability and reduced urinary platinum recovery. A significant sigmoidal relationship existed between ultrafilterable plasma AUC and the percentage of reduction in platelet count (r2 = 0.78). Nonlinear absorption was a limitation to this single-dose schedule of oral NM216; however, little non-haematological toxicity was seen at doses associated with myelosuppression and antitumour activity. Clinical studies of divided dose schedules using doses within the range of pharmacokinetic linearity (< or = 120 mg/m2) are now being investigated.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias/tratamento farmacológico , Compostos Organoplatínicos/administração & dosagem , Administração Oral , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Feminino , Humanos , Absorção Intestinal , Masculino , Pessoa de Meia-Idade , Compostos Organoplatínicos/efeitos adversos , Compostos Organoplatínicos/farmacocinética
18.
Cancer Chemother Pharmacol ; 33(6): 497-503, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-8137461

RESUMO

The preclinical toxicology and tissue platinum distribution of a series of six orally given antitumour platinum complexes [ammine/amine platinum(IV) dicarboxylates] with structural variations of their alicyclic amine (c-C5, c-C6 or c-C7), axial dicarboxylate (CH3, C3H7 or NHC2H5) or leaving substituents (Cl2 or OCOOCO) was studied in the mouse. Platinum tissue levels measured at 48 h after a single oral dose at 0.5 of the MTD were highest in the liver (6.0-19 micrograms/g) and second highest in the kidney (2.8-12 micrograms/g), and these levels were up to 5 times higher than those reported with equi-toxic doses of i.v. cisplatin and i.v. carboplatin. Platinum levels in the lung, heart, spleen, skin, skeletal muscle and brain were all < or = 3.1 micrograms/g at this dose level. Liver platinum levels measured at 2 h, 2 days, 6 days and 10 days after a single oral dose at the MTD ranged widely (from 15 to 109 micrograms platinum/g), were related to the number of carbon atoms in the axial dicarboxylate and alicyclic amine groups (r = 0.9389) and showed a diversity of time-course profiles. Elevations of plasma ALT activity were recorded with single oral doses of JM225 and JM256 at the MTD. Accumulation of platinum in the liver with repeated oral dosing weekly for 4 consecutive weeks at 0.5 of the MTD occurred with JM269 (3.3-fold increase, P < 0.05) and JM225 (2.4-fold increase, P < 0.05), and elevated plasma ALT activity (44 +/- 33 IU/l) was recorded with repeated oral doses of JM269. JM216 was selected from this series of analogues for further study on the basis of the elevated plasma ALT activity (JM225, JM256 and JM269), liver platinum accumulation (JM269 and JM225), poor activity against human ovarian carcinoma xenografts (JM291) or severe emetogenesis (JM221) of other examples. Following a single oral dose of JM216 at the MTD, transient reductions in the WBC (nadir, 1.6 x 10(9)/l, 2 days, 74% reduction), platelet count (nadir, 613 x 10(9)/l, 10 days, 33% reduction) and bone marrow cellularity (nadir, 0.5 x 10(7) nucleated cells/femur, 4 days, 75% reduction) were found, and these had recovered by 21 days after treatment. Jejunal mucosal disaccharidase activity following single MTDs indicated that small-intestinal mucosal damage was less severe for oral JM216 (nadir maltase activity, 68% +/- 16% of control, NS) than for i.v. cisplatin (nadir maltase activity).(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Antineoplásicos/farmacocinética , Antineoplásicos/toxicidade , Compostos Organoplatínicos/farmacocinética , Compostos Organoplatínicos/toxicidade , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Carboplatina/farmacocinética , Carboplatina/toxicidade , Cisplatino/farmacocinética , Cisplatino/toxicidade , Compostos Organoplatínicos/administração & dosagem , Fatores de Tempo , Distribuição Tecidual
19.
Oncol Res ; 11(6): 287-93, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10691031

RESUMO

Paclitaxel and cisplatin are associated with dose-limiting neurotoxicity that may result from their differing effects on microtubule stability in peripheral nerves. We hypothesized that such different actions of paclitaxel and cisplatin could be exploited to minimize their neurotoxicity by giving them in combination. Paclitaxel (9-18 micromol/kg/week or 7.7-15.4 mg/kg/week) and cisplatin (5-10 micromol/kg/week or 1.5-3 mg/kg/week) were given alone and in combination to female Wistar rats. Treatment was given once per week for a total of 7-10 weeks. Paclitaxel and cisplatin were given 24 h apart when they were given in combination. Changes in sensory nerve conduction velocity (SNCV) and dorsal root ganglia (DRG) morphology were measured. The nature of their interaction was analyzed using an isobologram. Their antitumor activity alone or in combination was also determined in C57B1/6 mice bearing colon 38 tumors. Reductions in SNCV occurred with paclitaxel alone (P = 0.009), cisplatin alone (P = 0.012), and cisplatin given 24 h before paclitaxel (P < 0.0001). In contrast, there was no significant change in SNCV with paclitaxel given 24 h before cisplatin (P = 0.11). An isobologram showed that the SNCV effects of the drug combinations were less than additive or antagonistic. Cisplatin-induced morphometric changes in DRG neurons were less marked when cisplatin was given with paclitaxel (P = 0.004). Concentrations of platinum in dorsal root ganglia, sural nerves, and sciatic nerves were not altered by giving paclitaxel before cisplatin. Tumor growth delays (TGD) were greater after treatment with paclitaxel (23.4 micromol/kg or 20 mg/kg) given 24 h before cisplatin (23.3 micromol/kg or 7 mg/kg) (TGD = 7.5 days) than after paclitaxel (23.4 micromol/kg or 20 mg/kg) (TGD = 2.0 days) or cisplatin (23.3 micromol/kg or 7 mg/kg) (TGD = 3.5 days) alone. Paclitaxel and cisplatin antagonized each other's neurotoxicity in Wistar rats. Combining cytotoxic agents with opposing effects on peripheral nerves has potential for minimizing neurotoxicity in patients.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Gânglios Espinais/efeitos dos fármacos , Condução Nervosa/efeitos dos fármacos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Cisplatino/administração & dosagem , Cisplatino/farmacocinética , Neoplasias do Colo/tratamento farmacológico , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Gânglios Espinais/química , Camundongos , Camundongos Endogâmicos C57BL , Paclitaxel/administração & dosagem , Paclitaxel/farmacocinética , Ratos , Ratos Wistar
20.
Drugs Aging ; 5(2): 85-95, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7981487

RESUMO

Compared with most solid tumours in humans, ovarian cancer may be considered a relatively chemosensitive disease. The platinum-based drug, cisplatin, has significantly enhanced long term survival (by more than 10%), though there remains scope for considerable improvement. Carboplatin is equiactive with, but substantially less toxic than, cisplatin (especially in terms of nephrotoxicity, effects on the gastrointestinal tract and neurotoxicity). Long term data are emerging from randomised trials that support the replacement of cisplatin by carboplatin in the first-line treatment of ovarian cancer. Iproplatin is also less toxic than cisplatin, but is more toxic and less active than carboplatin. A further advance in reducing patient morbidity associated with platinum-based chemotherapy may come from the first orally administrable platinum complex, JM216, which has recently been introduced into clinical trials. Current and future platinum drug discovery initiatives should focus on circumventing mechanisms of tumour resistance to cisplatin. Laboratory studies have identified 3 major mechanisms of resistance: reduced drug transport, enhanced intracellular detoxification (through glutathione and/or metallothioneins) and enhanced DNA removal (or tolerance) of platinum-DNA adducts (the long assumed critical lesions leading to cell kill). Platinum complexes based on the 1,2-diaminocyclohexane carrier ligand (such as oxaliplatin and tetraplatin) have recently entered clinical trials, having been shown to circumvent cisplatin resistance in murine leukaemia tumour models. Clinically, they have shown little evidence of activity in cisplatin-resistant disease and appear to cause severe neurotoxicity.


Assuntos
Antineoplásicos/uso terapêutico , Compostos Organoplatínicos/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Administração Oral , Carboplatina/uso terapêutico , Cisplatino/efeitos adversos , Cisplatino/uso terapêutico , Feminino , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA