Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cancer Chemother Pharmacol ; 69(5): 1145-54, 2012 May.
Article in English | MEDLINE | ID: mdl-22212299

ABSTRACT

PURPOSE: To determine the mass balance, excretion and metabolism of the small molecule flavonoid tumour vascular disrupting agent ASA404 in patients with advanced cancer. METHODS: Seven cancer patients were given a single dose of 3,000 mg [(14)C] ASA404 by intravenous infusion over 20 min prior to collection of samples of plasma, urine and faeces. Pharmacokinetic samples were analysed by HPLC, liquid scintillation counting, mass spectrometry, glusulase treatment and comparison to authentic standards. Descriptive pharmacokinetic parameters were generated by noncompartmental analysis. RESULTS: Mass balance was achieved (mean recovery of radioactivity in excreta = 86.9% of the dose) with balanced excretion between urine (mean recovery of radioactivity in urine = 53.9% of dose) and faeces (mean recovery of radioactivity in faeces = 33.3% of dose). ASA404 was eliminated as parent drug, three known metabolites (6-methylhydroxy-ASA404, ASA404 acyl glucuronide and 6-methylhydroxy-ASA404 acyl glucuronide) and two novel metabolites (an ASA404 dimer and an ASA404 dimer glucuronide conjugate). Unchanged ASA404 was the major radioactivity component detected in plasma within the first 24 h after dosing. At later time points, irreversibly protein bound ASA404 and all of the metabolites that had been detected in excreta contributed to total plasma radioactivity. CONCLUSION: This study defined the substantial excretion of ASA404, mainly as metabolites, in both urine (over half of the dose) and faeces (about one-third of the dose) after intravenous administration. Two novel metabolites were identified that were not reported by previous studies using nonradioactive techniques.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy , Aged , Antineoplastic Agents/therapeutic use , Carbon Radioisotopes , Chromatography, High Pressure Liquid/methods , Female , Humans , Infusions, Intravenous , Male , Mass Spectrometry , Middle Aged , Neoplasms/pathology , Protein Binding , Time Factors , Xanthones
2.
Br J Cancer ; 99(12): 2006-12, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19078952

ABSTRACT

ASA404 (5,6-dimethylxanthenone-4-acetic acid or DMXAA) is a small-molecule tumour-vascular disrupting agent (Tumour-VDA). This randomised phase II study evaluated ASA404 plus standard therapy of carboplatin and paclitaxel in patients with histologically confirmed stage IIIb or IV non-small cell lung cancer (NSCLC) not previously treated with chemotherapy. Patients were randomised to receive

Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Paclitaxel/therapeutic use , Xanthones/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carboplatin/pharmacology , Disease Progression , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Paclitaxel/pharmacology , Survival Rate , Treatment Outcome
3.
Neurotoxicology ; 28(6): 1092-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17686523

ABSTRACT

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.


Subject(s)
Cell Nucleolus/pathology , Ganglia, Spinal/pathology , Immunohistochemistry , Neurons, Afferent/pathology , Neurotoxicity Syndromes/pathology , Peripheral Nervous System Diseases/pathology , Activating Transcription Factor 3/analysis , Animals , Antineoplastic Agents, Phytogenic , Cell Size , Disease Models, Animal , Female , Ganglia, Spinal/chemistry , Ganglia, Spinal/physiopathology , H-Reflex , Neural Conduction , Neurons, Afferent/chemistry , Neuropeptide Y/analysis , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/physiopathology , Paclitaxel , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/physiopathology , Proto-Oncogene Proteins c-jun/analysis , Rats , Rats, Wistar , Reaction Time , Up-Regulation
4.
Cancer Chemother Pharmacol ; 56(4): 391-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15887017

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/toxicity , Ganglia, Spinal/drug effects , Neural Conduction/drug effects , Organoplatinum Compounds/toxicity , Peripheral Nervous System Diseases/chemically induced , Animals , Female , Oxaliplatin , Parvalbumins/isolation & purification , Peripheral Nervous System Diseases/pathology , Rats , Rats, Wistar , Substance P/isolation & purification
5.
Br J Cancer ; 88(12): 1942-7, 2003 Jun 16.
Article in English | MEDLINE | ID: mdl-12799641

ABSTRACT

Paclitaxel and oxaliplatin are promising drugs for combination trials but both induce peripheral neurotoxicity. To investigate this toxicity, 10-week-old female Wistar rats were given single intraperitoneal doses of paclitaxel and oxaliplatin, alone or in combination. Neurotoxicity was assessed by L5 dorsal root ganglion morphometry and H-reflex-related sensory nerve conduction velocity. Platinum concentrations in dorsal root ganglia and plasma were measured by inductively coupled plasma mass spectrometry. Dorsal root ganglion nucleolus size was significantly increased following single doses of paclitaxel of 10 and 20 mg kg(-1) at 24 h and 6 days (P<0.02). In contrast, dorsal root ganglion nucleolus size was significantly decreased following single doses of oxaliplatin ranging from 3 to 30 mg kg(-1) at time points ranging from 2 h to 14 days. Sensory nerve conduction velocity was altered after a single dose of oxaliplatin but not after paclitaxel. In combination with oxaliplatin, paclitaxel did not alter the plasma pharmacokinetics or dorsal root ganglion accumulation of oxaliplatin-derived platinum. However, prior paclitaxel inhibited oxaliplatin-induced reductions of dorsal root ganglion nucleolar diameter (P<0.02). Sensory nerve conduction velocity was reduced after oxaliplatin alone (P&<0.05) but unchanged when paclitaxel was given before oxaliplatin. In conclusion, paclitaxel induces nucleolar enlargement in dorsal root ganglion neurons after pharmacologically relevant doses in vivo and reduces oxaliplatin nucleolar damage and neurotoxicity.


Subject(s)
Ganglia, Spinal/drug effects , Organoplatinum Compounds/toxicity , Paclitaxel/pharmacology , Age Factors , Animals , Drug Therapy, Combination , Female , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Paclitaxel/administration & dosage , Rats
6.
Br J Cancer ; 85(8): 1219-25, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11710838

ABSTRACT

Platinum-based drugs are very useful in cancer therapy but are associated with neurotoxicity in the clinic. To investigate the mechanism of neurotoxicity, dorsal root ganglia of rats treated with various platinum drugs were studied. Cell body, nuclear and nucleolar dimensions of dorsal root ganglia sensory nerve cells were measured to determine morphological toxicity. Sensory nerve conduction velocity was measured to determine functional toxicity. After a single dose of oxaliplatin (10 mg kg(-1)), no significant change in nuclear and cell body diameter was seen but decreased nucleolar size was apparent within a few hours of treatment. Changes in nucleolar size were maximal at 24 hours, recovered very slowly and showed a non-linear dependence on oxaliplatin dose (r(2)= 0.99). Functional toxicity was delayed in onset until 14 days after a single dose of oxaliplatin but eventually recovered 3 months after treatment. Multiple doses of cisplatin, carboplatin, oxaliplatin, R, R-ormaplatin and S, S-ormaplatin were also associated with time-dependent reduction in nucleolar size. A linear correlation was obtained between the rate of change in nucleolar size during multiple dose treatment with the series of platinum drugs and the time taken for the development of altered sensory nerve conduction velocity (r(2)= 0.86;P< 0.024). Damage to the nucleolus of ganglionic sensory neurons is therefore linked to the neurotoxicity of platinum-based drugs, possibly through mechanisms resulting in the inhibition of rRNA synthesis.


Subject(s)
Antineoplastic Agents/toxicity , Cell Nucleolus/drug effects , Organoplatinum Compounds/toxicity , Animals , Carboplatin/toxicity , Cisplatin/toxicity , DNA Adducts/analysis , Dose-Response Relationship, Drug , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Ganglia, Spinal/physiology , Neural Conduction/drug effects , Oxaliplatin , Rats , Rats, Wistar
7.
J Inorg Biochem ; 85(2-3): 209-17, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11410241

ABSTRACT

A series of intercalator-tethered platinum(II) complexes PtLCl(2) have been prepared where L are the diamine ligands N-[2-[(aminoethyl)amino]ethyl]-9-aminoacridine-4-carboxamide, N-[3-[(2-aminoethyl)amino]propyl]-9-aminoacridine-4-carboxamide, N-[4-[(2-aminoethyl)amino]butyl]-9-aminoacridine-4-carboxamide and N-[5-[(aminoethyl)amino]pentyl]-9-aminoacridine-4-carboxamide and N-[6-[(aminoethyl)amino]hexyl]-9-aminoacridine-4-carboxamide. The activity of the complexes was assessed in the CH-1, CH-1cisR, 41M, 41McisR and SKOV-3 cell lines. The compounds with the shorter linker chain lengths are generally the most active against these cell lines and are much more toxic than Pt(en)C1(2). For example, for the n=2 compound the IC(50) values are 0.017 microM (CH-1), 1.7 microM (41M), 1.4 microM (SKOV-3) and the resistance ratios are 51 (CH-1cisR) and 1.6 (41McisR). For the untethered analogue Pt(en)C1(2) the IC(50) values are 2.5 microM (CH-1), 2.9 microM (41M), 45 microM (SKOV-3) and the resistance ratios are 2.8 (CH-1cisR) and 4.1 (41McisR). The very large differential in IC(50) values between the CH-1 and CH-1cisR pair of cell lines for the 9-aminoacridine-4-carboxamide tethered platinum complexes indicates that repair of platinum-induced DNA damage may be a major determinant of the activity of these compounds.


Subject(s)
Aminoacridines/pharmacology , Antineoplastic Agents/chemical synthesis , Organoplatinum Compounds/pharmacology , Aminoacridines/chemical synthesis , Aminoacridines/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Drug Resistance , Female , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Mice , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry , Tumor Cells, Cultured
8.
Expert Opin Investig Drugs ; 10(1): 119-28, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11116285

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclobutanes/therapeutic use , Organoplatinum Compounds/therapeutic use , Animals , Clinical Trials as Topic , Cyclobutanes/pharmacokinetics , Cyclobutanes/pharmacology , Humans , Organoplatinum Compounds/pharmacokinetics , Organoplatinum Compounds/pharmacology
9.
Cancer Chemother Pharmacol ; 46(5): 343-50, 2000.
Article in English | MEDLINE | ID: mdl-11127937

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Organometallic Compounds/pharmacokinetics , Organophosphorus Compounds/pharmacokinetics , Ovarian Neoplasms/metabolism , Phosphines/pharmacokinetics , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Chemical Phenomena , Chemistry, Physical , Female , Humans , Lipids/chemistry , Mice , Neoplasm Transplantation , Organogold Compounds , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Phosphines/chemistry , Phosphines/pharmacology , Protein Binding , Tumor Cells, Cultured
10.
J Biol Inorg Chem ; 5(6): 675-81, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11128994

ABSTRACT

Using assays based on the inhibition of restriction enzyme cleavage of plasmid and synthetic DNA, the complex (2-amino-2-methyl-3-butanone oxime)dichloroplatinum(II), [PtCl2(ambo)], has been shown to have an increased tendency for binding to adenine-rich sequences when compared to cis[PtCl2(NH3)2] (cisplatin). [PtCl2(ambo)] was found to form substantially fewer interstrand adducts than does cisplatin. The in vitro cytotoxicity of [PtCl2(ambo)] against a human bladder cancer cell line was determined and found to be more than two orders of magnitude lower than that of cisplatin, yet it was also found to be equally effective at passing into cells and binding to isolated DNA.


Subject(s)
Adenine/metabolism , Antineoplastic Agents/metabolism , DNA/drug effects , Organoplatinum Compounds/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Base Sequence , Cattle , DNA/chemistry , DNA/metabolism , Drug Design , Drug Screening Assays, Antitumor , Humans , Molecular Sequence Data , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacology , Tumor Cells, Cultured
11.
Br J Cancer ; 82(4): 966-72, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10732773

ABSTRACT

Previous work has shown platinum drugs to differ in their effects on the peripheral nervous system. To test whether their differential toxicity was due to differences in their partitioning into the peripheral nervous system, we correlated the hydrophobicity, reactivity, tissue accumulation and neurotoxicity of a series of eight platinum analogues. Neurotoxicity was detected by measuring sensory nerve conduction velocity (SNCV) in Wistar rats treated twice per week at the maximum tolerated dose. Tissue platinum concentrations were measured by inductively coupled plasma mass spectrometry. Hydrophobicity (log P) was measured using an octanol-aqueous shake-flask method. The half-life of platinum drug binding to plasma proteins in vitro was determined. The cumulative dose causing altered SNCV ranged from 15 to > 2050 micromol kg(-1). Ranking of the compounds by their neurotoxic potency in rats (oxaliplatin > R,R-(DACH)PtC4 > ormaplatin > S,S-(DACH)PtCl4 > S,S-(DACH)Pt oxalato > cisplatin > carboplatin > JM216) correlated with the frequency of neurotoxicity in patients (r> 0.99; P < 0.05). Ranking the compounds by their peripheral nerve accumulation was cisplatin > carboplatin > oxaliplatin > R,R-(DACH)PtCl4 = S,S-(DACH)PtCl4 and did not correlate with neurotoxicity. Log P ranged from - 2.53 to -0.16 but did not correlate with neurotoxicity. Log P correlated inversely with platinum accumulation in dorsal root ganglia (r2 = 0.99; P = 0.04), sural nerve (r2 = 0.85; P = 0.025), sciatic nerve (r2 = 0.98; P= 0.0012), spinal cord (r2 = 0.97, P= 0.018) and brain (r2 = 0.98, P= 0.001). Reactivity correlated with neurotoxicity potency in rats (r2 = 0.89, P = 0.0005) and with the frequency of neurotoxicity in patients (r2 = 0.99, P = 0.0002). The hydrophilicity of platinum drugs correlates with platinum sequestration in the peripheral nervous system but not with neurotoxicity. Differences in the reactivity of platinum complexes accounts for some of the variation in their neurotoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Organoplatinum Compounds/toxicity , Peripheral Nervous System Diseases/chemically induced , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Female , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/metabolism , Rats , Rats, Wistar , Stereoisomerism
12.
Cancer Invest ; 17(7): 479-85, 1999.
Article in English | MEDLINE | ID: mdl-10518192

ABSTRACT

The pharmacokinetics of vinblastine were studied in 16 patients with non-small cell lung cancer after a bolus intravenous dose of 3 mg/m2 given before or after cisplatin (100 mg/m2). Venous blood was collected at 0, 10, and 36 hr for analysis by radioimmunoassay. The mean plasma vinblastine concentration at 10 hr was similar when vinblastine was given before (4.8 ng/ml; 95% CI, 3.2-6.3) or after cisplatin (4.9 ng/ml; 95% CI, 2.7-7.1). Plasma vinblastine concentrations in patients given cisplatin were higher than previously reported in patients given vinblastine alone. Patients with plasma vinblastine concentrations less than 2.75 ng/ml at 10 hr experienced less severe neutropenia (37% fall in neutrophil count; 95% CI, 18-55) than those with levels greater than 2.75 ng/ml (69% fall in neutrophil count; 95% CI, 62-77). In conclusion, the pharmacokinetics of vinblastine predict the severity of neutropenia and may be altered when given in conjunction with cisplatin.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Vinblastine/pharmacokinetics , Aged , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/administration & dosage , Drug Administration Schedule , Female , Humans , Injections, Intravenous , Leukocyte Count/drug effects , Lung Neoplasms/drug therapy , Male , Middle Aged , Neoplasm Staging , Neutrophils/drug effects , Radioimmunoassay , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinblastine/therapeutic use
13.
Semin Oncol ; 26(3 Suppl 11): 13-8, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10458205

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Taxoids , Adult , Aged , Carcinoma, Non-Small-Cell Lung/secondary , Cisplatin/administration & dosage , Docetaxel , Drug Administration Schedule , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Paclitaxel/administration & dosage , Survival Analysis
14.
J Inorg Biochem ; 77(1-2): 105-10, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10626361

ABSTRACT

This paper reviews the neurotoxic side-effects associated with platinum drugs, experimental approaches to studying this toxicity and attempts to use neuroprotective agents in conjunction with platinum drugs. Platinum drugs differ in their neurotoxicity profiles in patients. The frequency, severity, mode of onset and reversibility of peripheral nerve toxicity varies between different platinum analogues. Animal models, primary cultures of dorsal root ganglia neurons and tumour cell-lines of neuronal origin are being used in attempts to identify potential treatments for platinum-induced neurotoxicity. To date, clinical trials have been hampered by the poor tolerance of neuroprotective treatments and failure to achieve reversal of platinum drug neurotoxicity with thiols, neurotrophic factors or calcium channel blockers.


Subject(s)
Antineoplastic Agents/adverse effects , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/prevention & control , Platinum Compounds/adverse effects , Animals , Antineoplastic Agents/toxicity , Calcium Channel Blockers/pharmacology , Carboplatin/adverse effects , Carboplatin/toxicity , Cisplatin/adverse effects , Cisplatin/toxicity , Disease Models, Animal , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Humans , Neurotoxicity Syndromes/etiology , Organoplatinum Compounds/adverse effects , Organoplatinum Compounds/toxicity , Oxaliplatin , Platinum Compounds/toxicity , Sulfhydryl Compounds/pharmacology
15.
Oncol Res ; 11(6): 287-93, 1999.
Article in English | MEDLINE | ID: mdl-10691031

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ganglia, Spinal/drug effects , Neural Conduction/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cisplatin/administration & dosage , Cisplatin/pharmacokinetics , Colonic Neoplasms/drug therapy , Drug Screening Assays, Antitumor , Female , Ganglia, Spinal/chemistry , Mice , Mice, Inbred C57BL , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Rats , Rats, Wistar
16.
J Clin Oncol ; 16(5): 1948-53, 1998 May.
Article in English | MEDLINE | ID: mdl-9586914

ABSTRACT

PURPOSE: Docetaxel (Taxotere, Rhone-Poulenc Rorer, Antony, France) and cisplatin are two of the most active single agents used in the treatment of non-small-cell lung cancer (NSCLC). A recently reported phase I study of the combination of docetaxel and cisplatin recommended a dose of 75 mg/m2 of both drugs every 3 weeks for subsequent phase II study. PATIENTS AND METHODS: Eligible patients were aged 18 to 75 years with a World Health Organization (WHO) performance status < or = 2 and life expectancy > or = 12 weeks, with metastatic and/or locally advanced NSCLC proven histologically or cytologically. Patients were not permitted to have received prior chemotherapy, extensive radiotherapy, or any radiotherapy to the target lesion and must have had measurable disease. Concurrent treatment with colony-stimulating factors (CSFs) or prophylactic antibiotics was not permitted. Docetaxel (75 mg/m2) in 250 mL 5% dextrose was given intravenously (i.v.) over 1 hour immediately before cisplatin (75 mg/m2) in 500 mL normal saline given i.v. over 1 hour in 3-week cycles. Premedication included ondansetron, dexamethasone, promethazine, and standard hyperhydration with magnesium supplementation. RESULTS: A total of 47 patients, two thirds of whom had metastatic disease, were entered onto this phase II study. The majority of patients were male (72%) and of good (WHO 0 to 1) performance status (85%). All 47 patients were assessable for toxicity and 36 were for response. Three patients were ineligible and eight (17%) discontinued treatment because of significant toxicity. In assessable patients, the overall objective response rate was 38.9% (95% confidence limits [CL], 23.1% to 56.5%), 36.1% had stable disease, and 25% progressive disease. On an intention-to-treat analysis, the objective response rate was 29.8%. Median survival was 9.6 months and estimated 1-year survival was 33%. Significant (grade 3/4) toxicities included nausea (26%), hypotension (15%), diarrhea (13%), and dyspnea mainly related to chest infection (13%). One patient experienced National Cancer Institute (NCI) grade 3 neurosensory toxicity after eight cycles. Grade 3/4 neutropenia was common and occurred in 87% of patients, but thrombocytopenia > or = grade 3 was rare (one patient). Significant (grade 3/4) abnormalities of magnesium levels were common (24%). Febrile neutropenia occurred in 13% of patients and neutropenic infection in 11%, contributing to two treatment-related deaths. No neutropenic enterocolitis or severe fluid retention was reported. CONCLUSION: Compared with other active regimens used in this setting, the combination of docetaxel and cisplatin in advanced NSCLC is an active regimen with a similar toxicity profile to other combination regimens.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Taxoids , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cisplatin/administration & dosage , Cisplatin/adverse effects , Disease Progression , Docetaxel , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Paclitaxel/analogs & derivatives , Survival Rate
17.
Met Based Drugs ; 5(4): 217-23, 1998.
Article in English | MEDLINE | ID: mdl-18475846

ABSTRACT

The in vitro cytotoxicities of a number of gold(I), silver(I) and copper(I) complexes containing chiral tertiary phosphine ligands have been examined against the mouse tumour cell lines P815 mastocytoma, B16 melanoma [gold(I) and silver(I) compounds] and P388 leukaemia [gold(I) complexes only] with many of the complexes having IC(50) values comparable to that of the reference compounds cis-diamminedichloroplatinum(ll), cisplatin, and bis[1,2-bis(diphenylphosphino) ethane]gold(I) iodide. The chiral tertiary phosphine ligands used in this study include (R)-(2-aminophenyl)methylphenylphosphine; (R,R)-, (S,S)- and (R(*),R(*))-1,2-phenylenebis(methylphenylphosphine); and (R,R)-, (S,S)- and (R(*),R(*))-bis{(2-diphenylphosphinoethyl)phenylphosphino}ethane. The in vitro cytotoxicities of gold(I) and silver(I) complexes containing the optically active forms of the tetra(tertiary phosphine) have also been examined against the human ovarian carcinoma cell lines 41M and CH1, and the cisplatin resistant 41McisR, CH1cisR and SKOV-3 tumour models. IC(50) values in the range 0.01 - 0.04 muM were determined for the most active compounds, silver(I) complexes of the tetra(tertiary phosphine). Furthermore, the chirality of the ligand appeared to have little effect on the overall activity of the complexes: similar IC(50) data were obtained for complexes of a particular metal ion with each of the stereoisomeric forms of a specific ligand.

18.
J Med Chem ; 40(22): 3508-15, 1997 Oct 24.
Article in English | MEDLINE | ID: mdl-9357517

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , DNA/drug effects , Organoplatinum Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cattle , Circular Dichroism , Crystallography, X-Ray , DNA/metabolism , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/metabolism , Stereoisomerism , Tumor Cells, Cultured
19.
J Clin Oncol ; 15(7): 2691-700, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9215842

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Administration, Oral , Adult , Aged , Antineoplastic Agents/adverse effects , Area Under Curve , Drug Administration Schedule , Female , Gastrointestinal Diseases/chemically induced , Hematologic Diseases/chemically induced , Humans , Male , Middle Aged , Neoplasms/blood , Neoplasms/drug therapy , Organoplatinum Compounds , Severity of Illness Index , Ultrafiltration
20.
Ann Oncol ; 8(5): 485-9, 1997 May.
Article in English | MEDLINE | ID: mdl-9233529

ABSTRACT

PURPOSE: This phase I study of paclitaxel and oral etoposide was performed to determine the safety of the combination in patients with advanced lung cancer who had received no prior chemotherapy, and to identify a dose for phase II testing. PATIENTS AND METHODS: Patients with locally advanced or metastatic non-small-cell lung cancer (NSCLC) or extensive small-cell lung cancer (SCLC), who had received no prior chemotherapy were treated with intravenous paclitaxel given as a three hour infusion (starting dose 100 mg/m2) and oral etoposide, 100 mg daily for five days. Two schedules of administration were used with the paclitaxel given on day 1 (schedule A) or day 5 (schedule B) of a 21 day cycle. RESULTS: Forty-nine patients were entered on the study, four of whom had SCLC. All patients were evaluable for toxicity. The maximum tolerated dose was paclitaxel 200 mg/m2 on day 1, in combination with oral etoposide 100 mg daily on days 1 to 5 (schedule A). The dose limiting toxicities were mucositis, myalgia, diarrhoea, and paraesthesiae. Using schedule B, myelosuppression, with febrile neutropenia was dose limiting at a paclitaxel dose of 160 mg/m2. Amongst the 45 patients with NSCLC there were three complete and eight partial responses (24%; 95% CI 13% 40%), while there was one complete response in the four patients with SCLC. CONCLUSION: Paclitaxel 200 mg/m2 on day 1, with oral etoposide 100 mg daily on days 1 to 5 can be administered safely, and is the recommended dose for phase II studies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Small Cell/drug therapy , Lung Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Etoposide/administration & dosage , Female , Humans , Male , Middle Aged , Paclitaxel/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...