Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Minerva Med ; 103(6): 415-29, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23229364

ABSTRACT

Symptomatic, severe aortic stenosis (AS) is associated with a dismal prognosis with conservative management only; the mortality rate is >50% at two years with medical treatment alone. In 1986, Cribier first introduced the percutaneous balloon aortic valvuloplasty (BAV) concept in patients with acquired severe AS. The initial enthusiasm surrounding this technique, touted as an alternative to surgical aortic valve replacement (SAVR) in older patients with AS, waned with subsequent large registries, which showed failure of the procedure to alter the natural history of calcific AS and its associated procedural morbidity. For many years BAV has been used as palliative treatment for short-term symptom relief in elderly, non-surgical patients. The timely surge in transcatheter aortic valve replacement (TAVR) rejuvenated and resurrected the dormant field of BAV. By its use to predilate the stenosed valve for easier delivery of the prosthesis, valvuloplasty now plays an integral role in the majority of TAVR procedures. BAV is successfully used as a bridge to SAVR and TAVR with better outcomes and is used as a standalone treatment for symptom relief in high-risk patients and for temporary stabilization of hemodynamically unstable patients. BAV can be used as a selection tool to determine if the patient will benefit from valve replacement if they have other comorbidities, such as severe pulmonary hypertension, severe lung disease, very poor ejection fraction, or frailty.


Subject(s)
Aortic Valve Stenosis/therapy , Aortic Valve , Balloon Valvuloplasty/methods , Aortic Valve Stenosis/physiopathology , Balloon Valvuloplasty/adverse effects , Balloon Valvuloplasty/trends , Calcinosis/therapy , Humans
2.
Minerva Cardioangiol ; 56(1): 89-105, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18432172

ABSTRACT

Although recent advances in percutaneous coronary interventions (PCI) have led to dramatic expansions in procedural complexity, bifurcation lesions (BL) remain a serious challenge for the interventionalist. Turbulent flow dynamics and high shear stress likely predispose coronary bifurcations to development of atherosclerotic plaques. These lesions comprise 15% to 20% of the total number of coronary interventions. When compared with non-BL interventions, BL interventions demonstrate lower procedural success rates, higher procedural costs, longer hospitalizations, and higher clinical and angiographic restenosis rates. The recent introduction of drug-eluting stents (DES) has resulted in lower incidences of target lesion/ vessel revascularization and reduction of main branch restenosis in this anatomic subset, when compared to historical bare metal stent (BMS) controls. Nonetheless, DES have not resolved the bifurcation PCI problem; and several techniques employing either 1 or 2 stents have emerged. Stenting of the main vessel with provisional side branch stenting seems to be the prevailing approach. While no definitive single BL-PCI technique has been identified, the optimal approach is likely lesion-specific. This paper reviews different treatment modalities for this complex lesion subset, with particular emphasis on the use of DES, as well as new potential therapeutic approaches.


Subject(s)
Angioplasty, Balloon, Coronary , Coronary Artery Disease/therapy , Coronary Vessels/pathology , Drug-Eluting Stents , Angioplasty, Balloon, Coronary/methods , Coronary Artery Disease/pathology , Humans , Randomized Controlled Trials as Topic , Treatment Outcome
3.
J Chemother ; 18(5): 514-24, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17127229

ABSTRACT

With a goal of identifying relations between gene expression and response (mucosal or pathological) or survival in esophageal cancer patients (stages II to IV) receiving oxaliplatin, 5-fluorouracil (5FU) and radiation, we measured in endoscopic primary tumor biopsies from 38 patients, the expression of seven genes (gammaGCS, gammaGT, MRP-2, ERCC-1, XPA, TS and DPD) prior to treatment, 1 week following oxaliplatin alone and at the end of the combined radio-chemotherapy cycle using real time QRT-PCR. A higher pretreatment level of XPA was related to shorter survival with a hazard ratio of 2.43 (90% confidence interval 1.09 to 5.43) using Cox regression modeling. However, multivariate analysis with a Cox model indicated low expression of XPA or TS and combined stages II and III had a higher probability of survival (for XPA: hazard ratio 3.0 and 90% C.I. of 1.3 to 6.9, with adjustment for stage included; for TS: hazard ratio is 1.98 with 90% C.I. of 0.94 to 4.20. The expression of TS, gammaGCS, ERCC-1 and MRP-2 declined from D 1 to the end of the cycle (p<0.05, sign test). A validation and further understanding of the findings need to be carried out in a larger study with a more homogeneous population of patients.


Subject(s)
Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Fluorouracil/pharmacology , Organoplatinum Compounds/pharmacology , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy/methods , Combined Modality Therapy , Endoscopy, Digestive System , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/radiotherapy , Female , Fluorouracil/administration & dosage , Gene Expression/drug effects , Humans , Laryngeal Mucosa/drug effects , Laryngeal Mucosa/metabolism , Male , Multidrug Resistance-Associated Protein 2 , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Radiation Dosage , Survival Analysis , Treatment Outcome
4.
Cancer Chemother Pharmacol ; 56(5): 447-54, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15947933

ABSTRACT

Two studies of irinotecan (CPT-11) followed 24 h later by an antimetabolite were conducted. The objectives of the studies were: (1) to determine whether the increase in S-phase in tumor cells seen 24 h after CPT-11 administration in animal studies is seen in advanced solid tumors in patients, (2) to determine the dose of CPT-11 required to produce this effect, (3) to compare two methods (immunohistochemistry, IHC, for cyclin A, and DNA flow cytometry, FC) for evaluating S-phase in tumor biopsies from patients, and (4) to establish the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) of CPT-11, given 24 h before gemcitabine (GEM, 1000 mg/m(2)). In one study CPT-11 was followed 24 h later by 5-fluorouracil (5-FU), 400 mg/m(2) per week for 4 weeks every 6 weeks. Tumor biopsies were obtained before and 24 h after CPT-11 administration before administration of 5-FU and assayed for S-phase by IHC for cyclin A and by FC. The starting dose of CPT-11 was 80 mg/m(2) per week with subsequent exploration of 40 and 60 mg/m(2) per week to establish the dose-effect relationship of the increase in tumor cells in S-phase. In the second study, CPT-11 was given 24 h before GEM 1000 mg/m(2) per week for 2 weeks every 3 weeks. Doses of 20-80 mg/m(2) were explored to establish the MTD and DLT and to study tumor cell S-phase in selected patients. CPT-11 80 mg/m(2) produced a mean increase in S-phase by IHC for cyclin A of 137%. Lesser increases were seen with 40 and 60 mg/m(2). CPT-11 followed 24 h later by 5-FU 400 mg/m(2) per week for 4 weeks was well tolerated. In the study of CPT-11 followed by GEM 1000 mg/m(2), 60 mg/m(2) of CPT-11 was the MTD.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Gastrointestinal Neoplasms/drug therapy , Respiratory Tract Neoplasms/drug therapy , S Phase/drug effects , Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Biopsy , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Cyclin A/analysis , Cyclin A/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacokinetics , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Gastrointestinal Neoplasms/pathology , Humans , Irinotecan , Male , Maximum Tolerated Dose , Respiratory Tract Neoplasms/pathology , Gemcitabine
5.
Cancer Epidemiol Biomarkers Prev ; 10(3): 269-72, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11303598

ABSTRACT

An N-acetylcysteine (NAC)/oltipraz (OLZ) combination was studied in healthy volunteer smokers who received daily NAC (1200 mg/day) and were randomized to weekly placebo (Arm A), OLZ 200 mg (Arm B), or 400 mg (Arm C). Treatment was for 12 weeks with follow-up at 16 weeks. The objective was to study toxicity and the modulation of pharmacodynamic end points. After treatment of 19 of a planned 60 subjects, (Arm A, six; Arm B, four; and Arm C, nine), the study was closed because of toxicity. Eight subjects failed to complete 12 weeks of drug administration, (Arm A, two, and Arm C, six). The most frequent side effects were gastrointestinal, fatigue, conjunctival irritation, and skin rash. Pharmacodynamic end points were measured pretreatment and 48 h after the dose of OLZ at weeks 1, 5, and 12 and 4 weeks after the end of treatment. Glutathione (GSH) was measured in plasma and in peripheral blood lymphocytes (PBLs). Other end points measured in PBLs were the enzyme activities of total glutathione-S-transferase (GST), GSTpi, and NAD(P)H:quinone oxidoreductase; and the mRNA expression of gamma-glutamylcysteine synthetase gammaGCS), GSTpi, and NAD(P)H:quinone oxidoreductase. GSH in PBLs, GST (total), and the mRNA of gammaGCS showed increases at some time points in some subjects. Most consistent was the mRNA of gammaGCS, which showed a > or = 30% increase at one or more time points in 11 of 19 subjects. Other end points were unchanged. We concluded that NAC/OLZ modulates some end points related to GSH but is too toxic for chemoprevention at the doses used.


Subject(s)
Acetylcysteine/administration & dosage , Acetylcysteine/toxicity , Gastrointestinal Diseases/chemically induced , Pyrazines/administration & dosage , Pyrazines/toxicity , Adult , Aged , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Fatigue/chemically induced , Female , Humans , Male , Middle Aged , Reference Values , Risk Assessment , Smoking , Thiones , Thiophenes
6.
Cancer Chemother Pharmacol ; 48(5): 398-406, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11761458

ABSTRACT

PURPOSE: We have previously reported that elevation of glutathione mediated by gamma-glutamyl transpeptidase is one mechanism of oxaliplatin resistance. This study explored other potential oxaliplatin resistance mechanisms with two aims: (1) to identify the differences between cisplatin and oxaliplatin in terms of drug accumulation, DNA-Pt adduct formation and repair, and (2) to determine whether defects in drug accumulation and enhanced repair of the DNA-Pt adduct contribute to oxaliplatin resistance. METHODS: The human ovarian carcinoma cell line A2780, an oxaliplatin-resistant variant A2780/C25 and a cisplatin-resistant variant A2780/CP along with an inherently cisplatin-resistant HT-29 colon carcinoma cell line were used in the study. The methods consisted of sulforhodamine-B assays, atomic absorption spectrophotometry and real-time quantitative RT-PCR. RESULTS: Significantly higher drug accumulation and DNA-Pt adduct formation were observed after exposure to cisplatin compared to after oxaliplatin in the parent A2780 cells and the oxaliplatin-resistant A2780/C25 cells. The DNA-Pt adduct formed after treatment with either drug was repaired with equal efficiency by all cell lines except A2780/CP, which repaired the DNA-cisplatin adduct more efficiently than the DNA-oxaliplatin adduct. Relative to the parent line, oxaliplatin-resistant A2780/C25 cells showed reduced Pt accumulation and DNA-Pt adduct levels following exposure to oxaliplatin, but only reduced accumulation after exposure to cisplatin. The cisplatin-resistant A2780/CP cells showed reduced accumulation and DNA-Pt adduct levels after exposure to cisplatin, but only reduced DNA-Pt adduct after exposure to oxaliplatin. In comparison to A2780 cells, the inherently cisplatin-resistant HT-29 cells showed lower accumulation and DNA-Pt adduct levels after exposure to cisplatin, but displayed no difference after exposure to oxaliplatin. An enhanced repair of the DNA-cisplatin adduct was observed only in A2780/CP cells relative to A2780 cells in an 8-h period. The steady-state levels of ERCC-1 mRNA, but not of XPA, were moderately elevated in the resistant cells. Exposure to either one of the drugs resulted in an induction of XPA in all the cell lines and of ERCC-1 in cisplatin-resistant cells. There was no relationship between the level of expression of the repair genes and the DNA-Pt adduct levels or repair. CONCLUSIONS: Relative to cisplatin a lower intracellular concentration and fewer DNA-Pt adducts are sufficient for oxaliplatin to exert its cytotoxicity. Resistance to oxaliplatin is mediated by similar mechanisms of reduced drug accumulation and DNA-Pt adduct formation as resistance to cisplatin. There is no clear evidence that enhanced repair is a mechanism of oxaliplatin resistance in the cell line (A2780/C25) studied here. The findings are suggestive of yet unidentified differences between the two drugs with respect to cellular uptake and/or efflux and repair of DNA-Pt adducts.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Binding Proteins , Endonucleases , Organoplatinum Compounds/pharmacology , DNA Adducts/analysis , DNA Repair , Drug Resistance, Neoplasm , HT29 Cells , Humans , Organoplatinum Compounds/pharmacokinetics , Oxaliplatin , Platinum/metabolism , Proteins/genetics , RNA, Messenger/analysis
7.
Methods Mol Med ; 39: 439-47, 2001.
Article in English | MEDLINE | ID: mdl-21340800

ABSTRACT

Heat shock protein 60 (HSP60, HSPD1) is a "chaperonin" that facilitates folding of nascent proteins into proper conformations (1). It is thought to play a critical role in the assembly, folding, and transport of proteins in the mitochondria. HSP60 also interacts with nascent cellular proteins to prevent their denaturation under heat stress (2). The HSP60 gene sequence is known and is highly conserved (3). Expression of the HSP60 gene has been associated with cisplatin resistance in several preclinical model systems and in ovarian carcinoma patients (1-6); we quantitated HSP60 mRNA expression in preclinical human ovarian and bladder carcinoma models.

8.
Clin Cancer Res ; 6(4): 1314-21, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10778956

ABSTRACT

Depletion of cellular glutathione (GSH) enhances the efficacy of many anticancer agents in preclinical systems. Limited published data showing depletion of GSH in vitro and in patients by ifosfamide and/or mesna provided the rationale for a Phase I trial. Ifosfamide and mesna were infused over 24 and 36 h, respectively, at equal daily doses; carboplatin was given after ifosfamide to a target plasma area under the curve of 4 mg x min x ml(-1). Plasma and peripheral WBC thiols were quantitated by high-performance liquid chromatography. The dose of ifosfamide was escalated from 2 to 8 g/m2; the maximum tolerated dose was 6 g/m2. Significant depletion in plasma cysteine and homocysteine, precursors for GSH synthesis, was observed (maximum, 95% to >99% at 8 g/m2). Plasma mesna and cysteine/ homocysteine levels were inversely correlated; nadir levels of cysteine/homocysteine were maintained for several hours after ifosfamide infusion had stopped and while mesna infusion was continuing. In vitro coincubation experiments confirmed that mesna reduces these thiols from disulfides to sulfhydryls, which are readily cleared, as evidenced by the significantly increased rate of excretion of cysteine in urine. In contrast, ifosfamide/mesna treatment caused a moderate depletion of plasma GSH in only 60% of the patients, with a nadir at 24 h and recovery immediately after the end of ifosfamide infusion. The GSH depletion in these patients was not dose related. The profile of GSH recovery in plasma after ifosfamide and the fact that mesna could not reduce GSH disulfides in vitro suggest that the observed GSH depletion in plasma in 60% of the patients may be related to direct reactions of GSH with ifosfamide metabolites and/or mesna. Our results indicate that mesna is a modulator of GSH precursors and that a prolonged infusion of mesna may be required to achieve GSH precursor starvation and the consequent GSH depletion in cells.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Glutathione/drug effects , Ifosfamide/therapeutic use , Leukocytes/drug effects , Mesna/pharmacology , Sulfhydryl Compounds/blood , Acetaldehyde/analogs & derivatives , Acetaldehyde/blood , Antineoplastic Agents, Alkylating/pharmacokinetics , Cysteine/blood , Cysteine/drug effects , Cysteine/urine , Dose-Response Relationship, Drug , Glutathione/blood , Homocysteine/blood , Homocysteine/drug effects , Humans , Ifosfamide/pharmacokinetics , Infusions, Intravenous , Leukocytes/metabolism , Mesna/blood , Neoplasms/blood , Neoplasms/drug therapy , Oxidation-Reduction , Protective Agents/pharmacology
9.
Cancer Invest ; 17(7): 463-7, 1999.
Article in English | MEDLINE | ID: mdl-10518189

ABSTRACT

Patients with stage IIIB breast carcinoma represent only a small proportion of women with breast cancer in western countries but may constitute up to 50% of cases in underdeveloped countries. The prognosis remains poor despite aggressive treatment. Nineteen patients (11 with inflammatory breast carcinoma) received at least three courses of neoadjuvant chemotherapy of methotrexate, vinblastine, adriamycin, cisplatin, and folinic acid (MVAC/FA) followed by mastectomy. Six months of cyclophosphamide, methotrexate, and 5-fluorouracil were given after surgery. Radiation therapy followed chemotherapy. Seventy percent of patients achieved complete and 14% partial response after MVAC/FA chemotherapy alone. Eleven patients (58%) survived 5 years, and 30% survived at least 8 years. The addition of cisplatin in combination chemotherapy used as first-line treatment for stage IIIB breast carcinoma was well tolerated, resulted in higher response rates, and appeared to have an effect on overall survival.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/mortality , Cisplatin/administration & dosage , Doxorubicin/administration & dosage , Drug Administration Schedule , Female , Fluorouracil/administration & dosage , Humans , Leucovorin/administration & dosage , Methotrexate/administration & dosage , Middle Aged , Neoadjuvant Therapy , Neoplasm Staging , Prognosis , Survival Analysis , Vinblastine/administration & dosage
10.
Cancer Chemother Pharmacol ; 41(2): 167-70, 1998.
Article in English | MEDLINE | ID: mdl-9443631

ABSTRACT

PURPOSE: To establish the maximum tolerated dose (MTD), dose-limiting and other major toxicities and the major pharmacokinetic parameters of a 10-day infusion of the nonclassical antifolate Thymitaq. METHODS: The drug was given by 10-day infusion via a portable pump. The starting dose was 286 mg/m2 per day with escalation to 572 and 716 mg/m2 per day. Thymitaq in plasma was assayed by a validated isocratic reverse-phase HPLC assay with detection at 273 nm. RESULTS: The dose of 716 mg/m2 per day x 10 was considered too high as none of three patients completed a 10-day infusion and two of three developed grade IV myelotoxicity. At 572 mg/m2 per day three of four patients completed a 10-day infusion. Dose-limiting myelosuppression was seen in one of four but owing to a high incidence of thrombotic phenomena, no further patients were added. CONCLUSION: Continuous 10-day infusions of Thymitaq should be limited to low doses until further studies can be done.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Neoplasms/drug therapy , Quinazolines/administration & dosage , Adult , Aged , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/pharmacokinetics , Area Under Curve , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Humans , Infusions, Intravenous , Middle Aged , Quinazolines/adverse effects , Quinazolines/blood , Quinazolines/pharmacokinetics
11.
Eur J Cancer ; 34(10): 1543-8, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9893625

ABSTRACT

Paclitaxel and carboplatin have nonoverlapping toxicities with a broad range of clinical activity. The combination of escalating dose paclitaxel and carboplatin dosed to a fixed area under the curve (AUC) was explored in a series of phase I studies. 76 patients were treated with paclitaxel over three hours followed by a 30 min carboplatin infusion, dosed by the Calvert formula to a target AUC of 4.0 or 4.5 mg/min/ml-1. The maximum tolerated dose of paclitaxel was 270 to 290 mg/m2, with a dose limiting toxicity of peripheral sensory neuropathy. Activity was seen in lung cancer, with a paclitaxel dose at or above 230 mg/m2. Neuropathy correlated with paclitaxel AUC due to nonlinear pharmacokinetics at higher doses. Ongoing studies include the use of amifostine as a neuroprotectant and phase II studies of the paclitaxel/carboplatin regimen in head and neck cancer, small cell lung cancer and sarcomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Adult , Aged , Carboplatin/administration & dosage , Carboplatin/adverse effects , Clinical Trials, Phase I as Topic , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects
12.
Semin Oncol ; 24(4 Suppl 12): S12-138-S12-143, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9331139

ABSTRACT

The combination of paclitaxel (Taxol; Bristol-Myers Squibb Company, Princeton, NJ) given by 3-hour infusion followed by carboplatin infused over 30 minutes has been evaluated in a series of phase I studies and is currently being explored in a phase II study in patients with limited- and extensive-stage small cell lung cancer. Pharmacokinetic measurements were performed at all dose levels in the phase I studies, in which the use of granulocyte colony-stimulating factor in previously treated patients enabled more than twice the dose of paclitaxel to be given with low to moderate doses of carboplatin (dosed to a target area under the concentration-time curve of 4.0 mg x min x mL[-1]). Treatment-naive patients tolerated high paclitaxel doses (270 mg/m2) with carboplatin (dosed to a target area under the curve of 4.5 mg x min x mL[-1]) without granulocyte colony-stimulating factor support. Twenty-three patients (including previously treated and untreated) with non-small cell lung cancer were entered at a variety of paclitaxel doses in the phase I studies. At 100 to 205 mg/m2 paclitaxel, none of nine treated patients responded; at 230 to 290 mg/m2, four (29%) of 14 responded. In the phase II study of paclitaxel 250 mg/m2 in previously untreated patients with small cell lung cancer, two of five evaluable patients with extensive-stage disease have shown a partial response. In a preliminary analysis of the pharmacodynamics of paclitaxel in relation to neurotoxicity (dose limiting in two of three phase I studies), neurotoxicity correlated with the total dose of paclitaxel, the area under the curve, and the peak paclitaxel concentration, but not with the length of time plasma paclitaxel levels remained above 0.05 micromol/L. These correlations were not strong, however, and analysis of these data is ongoing.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Small Cell/drug therapy , Lung Neoplasms/drug therapy , Paclitaxel/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carboplatin/administration & dosage , Carboplatin/pharmacokinetics , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Humans , Paclitaxel/pharmacokinetics
13.
Clin Cancer Res ; 3(5): 793-8, 1997 May.
Article in English | MEDLINE | ID: mdl-9815751

ABSTRACT

Glutathione (GSH) levels were measured in 13 human tumor cell lines derived from carcinomas of the bladder, ovary, and colon and from melanoma and glioblastoma. High levels were found in four of five bladder cell lines. The average GSH concentration in the bladder cell lines was approximately 6-fold higher than in the non-bladder cell lines. Because this difference suggested the possibility of elevation of GSH in urothelial neoplasia, we measured GSH in bladder tumor tissue from patients with transitional cell carcinoma (TCC) of the bladder (Group I, n = 17). GSH was also measured in two types of control tissues: (a) nontumor bladder tissue from patients with TCC or a history of TCC of the bladder (Group II, n = 23); and (b) bladder tissue from patients without bladder cancer (Group III, n = 14). Thirteen sets of paired specimens of tumor and nontumor bladder tissue from the same patient were evaluated. The tissues were flash-frozen, and GSH was measured after histological assessment of the same samples. Free and total GSH (free + mixed disulfides) were measured by a high-performance liquid chromatography assay with fluorescence detection and expressed as nanomoles/mg protein. The mean free GSH (+/- SD) for groups I, II, and III was 32.0 +/-18.7, 17.3 +/- 11.4, and 9.3 +/- 4.0, respectively, and the mean total GSH was 45.9 +/- 32.5, 23.7 +/- 17.1, and 12.2 +/- 6.7. The respective differences between groups (I and II, I and III, and II and III) were statistically significant for both free and total GSH (Ps ranging from <0.0001 to

Subject(s)
Carcinoma, Transitional Cell/metabolism , Glutathione/metabolism , Urinary Bladder Neoplasms/metabolism , Brain Neoplasms/chemistry , Carcinoma, Transitional Cell/chemistry , Carcinoma, Transitional Cell/pathology , Carcinoma, Transitional Cell/surgery , Female , Glioblastoma/chemistry , Glutathione/analysis , Humans , Melanoma/chemistry , Neoplasm Staging , Ovarian Neoplasms/chemistry , Tumor Cells, Cultured , Urinary Bladder/chemistry , Urinary Bladder/pathology , Urinary Bladder Neoplasms/chemistry , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery , Urothelium/chemistry , Urothelium/pathology
14.
J Clin Oncol ; 15(2): 766-72, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9053503

ABSTRACT

PURPOSE: Etoposide has schedule-dependent cytotoxic activity, and clinical resistance may be overcome with prolonged low-dose therapy. Oral bioavailability is variable, and protracted intravenous administration is limited by water insolubility, which requires large infusion volumes. Etoposide phosphate (EP) is a water-soluble prodrug that is rapidly converted in vivo to etoposide, and may be administered in concentrated solution. A phase I study was conducted to determine the toxicity, pharmacokinetics, and pharmacodynamics of EP administered as a protracted venous infusion in the ambulatory setting. METHODS: Twenty-three patients with advanced cancer were treated with a continuous infusion of EP using ambulatory pumps for 6 weeks followed by a 2-week rest. Cohorts were treated with EP at 10, 20, 25, and 30 mg/m2/d. Steady-state plasma etoposide levels (Css) and stability of EP in infusion pumps were measured using high performance liquid chromatography (HPLC). RESULTS: Myelosuppression, mucositis, and fatigue were dose-limiting. The maximum-tolerated dose (MTD) of EP was 20 mg/m2/d. The mean Css (+/- SD) of etoposide were 0.67 +/- 0.25, 1.14 +/- 0.24, 1.38 +/- 0.64, and 2.19 +/- 0.52 microg/mL at daily EP doses of 10, 20, 25, and 30 mg/m2, respectively. Neutropenia correlated with Css (r = 0.65, P = .008). EP was stable in infusion pumps for at least 7 days. Partial responses were observed in patients with hepatoma and non-small-cell lung cancer (one each). CONCLUSION: EP may be conveniently and safely administered as a low-volume protracted venous infusion in the ambulatory setting. Cytotoxic plasma concentrations of etoposide are obtained at the MTD. The pharmacodynamic relationships observed suggest the possibility of pharmacologically based dosing of EP.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Etoposide/analogs & derivatives , Neoplasms/drug therapy , Organophosphorus Compounds/pharmacokinetics , Organophosphorus Compounds/therapeutic use , Adult , Aged , Ambulatory Care , Antineoplastic Agents/adverse effects , Biological Availability , Etoposide/adverse effects , Etoposide/pharmacokinetics , Etoposide/therapeutic use , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/blood , Organophosphorus Compounds/adverse effects , Treatment Outcome
15.
Invest New Drugs ; 15(3): 227-34, 1997.
Article in English | MEDLINE | ID: mdl-9387045

ABSTRACT

The objectives of this study were to determine the dose limiting toxicity (DLT) and other major toxicities, the maximum tolerated dose (MTD) and the human pharmacokinetics of N1N11 diethylnorspermine (DENSPM), a new polyamine analog which in experimental systems inhibits the biosynthesis of intracellular polyamines and promotes their degradation by inducing the enzyme spermine/spermidine N-acetyl transferase. These objectives were incompletely achieved because of the occurrence of an unusual syndrome of acute central nervous system toxicity which forms the basis of the present report. Fifteen patients with advanced solid tumors were entered into a phase I study of DENSPM given by a 1 h i.v. infusion every 12 h for 5 days (10 doses). The starting dose was 25 mg/m2/day (12.5 mg/m2/dose) with escalation by a modified Fibonacci search. Doses of 25 and 50 mg/m2/day were tolerated with only minor side effects of facial flushing, nausea, headache and dizziness (all grade I). At doses of 83 and 125 mg/m2/day, a symptom complex of headache, nausea and vomiting, unilateral weakness, dysphagia, dysarthria, numbness, paresthesias, and ataxia, was seen in 3 patients, one after 2 courses of 83 and 2 after 1 course of 125 mg/m2/day. This syndrome occurred after drug administration was complete and the patients had returned home. Lesser CNS toxicity was seen in 2 other patients at lower daily doses. Preliminary pharmacokinetics of DESPM measured in plasma by HPLC in 8 patients showed linearity with dose and a rapid plasma decay with a t1/2 of 0.12 h. We conclude that great caution is warranted in administering DENSPM on this schedule at doses of > or = 83 mg/m2/day.


Subject(s)
Antineoplastic Agents/adverse effects , Central Nervous System Diseases/chemically induced , Neoplasms/drug therapy , Spermine/analogs & derivatives , Adenocarcinoma/drug therapy , Adult , Aged , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Biological Availability , Colonic Neoplasms/drug therapy , Female , Half-Life , Humans , Lung Neoplasms/drug therapy , Male , Melanoma/drug therapy , Metabolic Clearance Rate , Middle Aged , Spermine/adverse effects , Spermine/pharmacokinetics
16.
Cancer Chemother Pharmacol ; 40(1): 38-44, 1997.
Article in English | MEDLINE | ID: mdl-9137527

ABSTRACT

Previous studies from our laboratory have indicated that glutathione (GSH) may affect the cytotoxicity of iproplatin to a greater extent than four other platinum agents tested including cisplatin. Therefore we studied the effect of GSH depletion by buthionine sulfoximine (BSO) on the cytotoxicity of iproplatin and cisplatin in a human melanoma cell line SK-MEL-2. Depletion of GSH was dependent on the concentration and time of incubation with BSO. BSO (100 microM) depleted GSH by 85% at 24 h and by 91% at 48 h. BSO (10 to 100 microM) by itself was not cytotoxic to SK-MEL-2 cells. At 85% depletion of GSH, cytotoxicity of iproplatin was increased by a factor of > 7 and that of cisplatin by < 2. These results confirm the previous finding that GSH interferes with the cytotoxicity of iproplatin to a significantly greater extent than that of cisplatin. Equitoxic IC65 and IC90 values of cisplatin (2 microM and 5 microM) or iproplatin (25 microM and 50 microM) had no effect on the intracellular GSH levels in SK-MEL-2 cells. Also, depletion of GSH by BSO had no effect on the accumulation of platinum from either cisplatin or iproplatin in this cell line. Our results suggest that the effect of GSH on the cytotoxicity of cisplatin and iproplatin in this cell line was not a consequence either of differences in GSH-Pt conjugate formation, or of differences in platinum accumulation induced by GSH depletion. GSH may have modulated the cytotoxicity of these platinum complexes by other means such as effects on DNA repair, apoptosis, free radical scavenging or through other yet unidentified mechanisms.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Glutathione/physiology , Organoplatinum Compounds/pharmacology , Buthionine Sulfoximine/pharmacology , Cell Survival/drug effects , Glutathione/analysis , Humans , Melanoma/pathology , Tumor Cells, Cultured
17.
Cancer Lett ; 119(1): 63-70, 1997 Oct 28.
Article in English | MEDLINE | ID: mdl-18372523

ABSTRACT

Heat shock protein 60 (HSP60) participates in protein assembly, folding and transport. Increased HSP60 mRNA expression is associated with cisplatin resistance in some in vitro models and with shorter survival among ovarian cancer patients. HSP60 mRNA expression was quantitated in three independent model systems by reverse-transcription PCR (A2780 human ovarian carcinoma cell line and sublines selected for cisplatin or oxaliplatin resistance in vitro and also in the UCRU-BL13 human bladder carcinoma cell line and a cisplatin-resistant subline). Increased HSP60 mRNA expression was observed in all resistant sublines (range 2.5-15 fold), correlated with relative resistance to cisplatin and oxaliplatin. No differences in HSP60 gene copy number were apparent in resistant sublines. These data provide further evidence of a strong association between in vitro resistance to platinum compounds and increased HSP60 mRNA expression.


Subject(s)
Antineoplastic Agents/pharmacology , Chaperonin 60/biosynthesis , Drug Resistance, Neoplasm/physiology , Ovarian Neoplasms/metabolism , Platinum Compounds/pharmacology , Urinary Bladder Neoplasms/metabolism , Cell Line, Tumor , Chaperonin 60/genetics , Female , Gene Dosage , Gene Expression , Humans , Ovarian Neoplasms/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Urinary Bladder Neoplasms/genetics
18.
Semin Oncol ; 23(6 Suppl 16): 26-31, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007117

ABSTRACT

In preparation for the design of phase II studies in lung cancer, low-dose carboplatin, fixed at a target area under the concentration-time curve (AUC) of 4.0 or 4.5 mg x min/mL, has been combined with escalating doses of paclitaxel (Taxol; Bristol-Myers Squibb Company, Princeton, NJ) in a series of studies to establish the maximum tolerated dose of the combination. In patients who had received prior chemotherapy, the maximum tolerated paclitaxel dose was 135 mg/m2 (carboplatin target AUC 4.0); the dose-limiting toxicity was febrile neutropenia. Without granulocyte colony-stimulating factor support in chemotherapy-naive patients (carboplatin target AUC 4.5), and with granulocyte colony-stimulating factor in chemotherapy-pretreated patients, the current paclitaxel dose is 290 mg/m2. The maximum tolerated dose has not been defined. In a study in which paclitaxel was given by 1-hour infusion with carboplatin (target AUC 4.5), a 205 mg/m2 dose was poorly tolerated. No evidence of pharmacokinetic interactions between paclitaxel and carboplatin was found. Twenty-one evaluable patients with lung cancer have been treated to date. There have been two partial responses, one minor response, and 10 patients with stable disease at paclitaxel doses of 100 to 270 mg/m2.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Paclitaxel/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/toxicity , Carboplatin/administration & dosage , Drug Tolerance , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Male , Paclitaxel/pharmacokinetics , Paclitaxel/toxicity , Treatment Outcome
19.
Cancer Lett ; 105(1): 5-14, 1996 Jul 19.
Article in English | MEDLINE | ID: mdl-8689632

ABSTRACT

Elevation of glutathione (GSH) is commonly observed in cellular resistance to a number of anticancer agents. Most frequently reported change in GSH metabolism that is associated with the elevated GSH levels is increased mRNA expression and activity of gamma-glutamyl cysteine synthetase (gamma GCS), the first enzyme of the GSH biosynthetic pathway. We have isolated sublines of the A2780 ovarian carcinoma cell line (C10 and C25) that are 8- and 12-fold resistant to oxaliplatin by repeatedly exposing the cells to increasing concentrations of the platinum agent. The GSH levels in C10 and C25 cell sublines are 3.1- and 3.8-fold higher than the parent A2780 cell line. The mRNA levels and activities for gamma GCS and that for gamma-glutamyl transpeptidase (gamma GT), the GSH salvage pathway enzyme, were measured in these cells. The mRNA for gamma GT and gamma GCS were measured by RT-PCR, with quantitation of the PCR product by HPLC; mRNA levels are expressed as ratios to beta-actin mRNA, used as an endogenous standard. GSH and gamma GCS activity were measured by HPLC assays and gamma GT activity by a colorimetric assay. The increase in GSH in C10 and C25 was associated with an elevation in gamma GT mRNA (2.5- and 8-fold) and gamma GT activity (2.7- and 2.8-fold). No changes were observed in gamma GCS mRNA levels or activity. The data indicate that alterations in GSH metabolism leading to elevations in cellular GSH in A2780 ovarian carcinoma cells selected for low levels of resistance to oxaliplatin are mediated by gamma GT, the "salvage' pathway, rather than an increase in GSH biosynthesis.


Subject(s)
Carcinoma/metabolism , Glutathione/metabolism , Organoplatinum Compounds/toxicity , Ovarian Neoplasms/metabolism , gamma-Glutamyltransferase/metabolism , Antineoplastic Agents/toxicity , Base Sequence , DNA Primers/chemistry , Drug Resistance , Female , Gene Expression Regulation, Neoplastic/drug effects , Glutamate-Cysteine Ligase/metabolism , Humans , Molecular Sequence Data , Oxaliplatin , RNA, Messenger/genetics
20.
Oncol Res ; 8(10-11): 415-23, 1996.
Article in English | MEDLINE | ID: mdl-9114434

ABSTRACT

Elevation of glutathione (GSH) is widely observed in cellular resistance to platinum agents. Our previous studies have shown that sublines of human ovarian carcinoma cell line A2780, which exhibited low levels of resistance to oxaliplatin, showed elevated steady state levels of mRNA and activity of gamma-glutamyl transpeptidase (gamma-GT, EC 2.3.2.2), but not of gamma-glutamylcysteine synthetase (gamma-GCS, EC 6.3.2.2) [El-akawi et al., Cancer Lett. 105:5-14; 1966]. To understand this phenomenon better, we have studied the effect of single exposures of oxaliplatin or cisplatin on the mRNA expression of gamma-GT and gamma-GCS in A2780 cells. The mRNAs of gamma-GT and gamma-GCS were measured by reverse transcriptase PCR, with quantitation of the PCR product by HPLC; mRNA levels are expressed as ratios to beta-actin mRNA, used as an endogenous standard. GSH was measured by HPLC. The gamma-GT activity was measured by a colorimetric assay. Single exposures of cells to oxaliplatin induced a time- and concentration-dependent increase in the mRNA of gamma-GT, but not of gamma-GCS. Cisplatin also induced an elevation in gamma-GT mRNA, but to a lower degree. The gamma-GT enzyme activity increased corresponding to the elevation in mRNA expression. The gamma-GT-induced cells showed an increase in cellular GSH when incubated in medium containing GSH. The data suggest that a) single, brief exposures to pharmacologically relevant concentrations of platinum complexes induce elevation in mRNA of gamma-GT, b) elevation in gamma-GT mRNA translates into elevated gamma-GT activity and increase in GSH salvage, and c) the degree of induction of gamma-GT mRNA differs between platinum complexes.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Organoplatinum Compounds/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , RNA, Messenger/biosynthesis , gamma-Glutamyltransferase/biosynthesis , Base Sequence , Cell Division/drug effects , Drug Resistance, Neoplasm , Enzyme Induction/drug effects , Female , Glutamate-Cysteine Ligase/biosynthesis , Glutamate-Cysteine Ligase/metabolism , Glutathione/biosynthesis , Glutathione/metabolism , Humans , Molecular Sequence Data , Oxaliplatin , Tumor Cells, Cultured , gamma-Glutamyltransferase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...