Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Ann Oncol ; 12(8): 1067-73, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11583187

ABSTRACT

BACKGROUND: To determine the cardiotoxicity of paclitaxel (T) plus doxorubicin (A) combination therapy in women with advanced breast cancer. To define a dose range of A for use in AT. PATIENTS AND METHODS: The effect of cumulative A dose on risk of congestive heart failure (CHF) and alterations of myocardial contractility (left ventricular ejection fraction [LVEF] decrease > or = 20% or to <50%) was estimated from pooled data from 10 trials of AT. RESULTS: Thirty-one of 657 patients (4.7%) developed CHF at a median of 6.6 months (range 0.3-24.6) after initiation of AT. CHF was stabilized in 29 patients at a median of 17.3 months after diagnosis (range 4.1-31.2 months). The risk of developing CHF was < or = 5% at a total A dose < or = 380 mg/m2. In patients who received a total A dose > 440 mg/m2, the incidence of CHF was >25% but similar to that of A monotherapy. The risk of CHF was similar in women receiving AT or A monotherapy at a dose < or = 380 mg/m2 (2%-3%). LVEF progressively decreased in patients who received AT, especially at a cumulative A dose > 380 mg/m2. LVEF decreases were more frequent in patients who later developed CHF, but the majority of CHF patients did not experience LVEF alterations prior to symptoms. LVEF recovered after discontinuation of A in 25 of 67 women who developed LVEF < 50%. CONCLUSION: The reported cardiac effects are consistent with anthracycline-related cardiotoxicity. AT is associated with a cardiac risk similar to that of A monotherapy up to a cumulative A dose of 340-380 mg/m2.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/drug therapy , Heart Failure/chemically induced , Adult , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/physiopathology , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Female , Humans , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Retrospective Studies , Stroke Volume/drug effects , Ventricular Function, Left/drug effects
2.
Anticancer Drugs ; 10(9): 821-7, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10587292

ABSTRACT

BMS-181174, a new mitomycin C (MMC) analog, showed more activity than the parent compound in tumor xenografts. In a phase I study with a 5-30 min slow bolus administration, hematologic and vascular toxicity were observed as major side effects. A prolonged infusion was suggested to circumvent the vascular toxicity. In this phase I study BMS-181174 was administered as a 6 h infusion every 4 weeks; the doses studied were 3.2, 6.4, 11.5, 19.0, 32.0, 50.0, 75.0 and 100 mg/m2. Twenty-eight patients were enrolled in the study, the majority with colorectal cancer. Hematologic side effects consisted of thrombocytopenia, and mild leuko- and granulocytopenia. The most distressing non-hematologic side effect was vascular toxicity consisting of phlebosclerosis and phlebitis, becoming dose limiting at 100 mg/m2. One patient developed a hemolytic uremic syndrome at a cumulative dose of 350 mg/m2. Pharmacokinetic data are available for 24 patients. The AUC ranged from 3.35 to 41.49 (microg x h/ml) and was highly correlated with the dose administered (r = 0.83). The kinetics appeared to be linear. One patient with metastatic colon cancer had a partial response of liver metastases. BMS-181174 is a MMC analog with a toxicity profile comparable to that of the parent compound. As doses above 50 mg/m2 are complicated by vascular toxicity precluding multiple administrations, further exploration of BMS-181174 will not be performed.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Hematologic Diseases/chemically induced , Mitomycin , Mitomycins , Mitomycins/administration & dosage , Mitomycins/adverse effects , Neoplasms/drug therapy , Vascular Diseases/chemically induced , Adult , Aged , Antineoplastic Agents, Alkylating/pharmacokinetics , Area Under Curve , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Mitomycins/pharmacokinetics , Neoplasm Staging , Neoplasms/blood , Neoplasms/pathology
3.
J Clin Oncol ; 17(6): 1876-83, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10561228

ABSTRACT

PURPOSE: Liposomal anthracyclines are the present standard treatment for advanced AIDS-related Kaposi's sarcoma (KS). No effective therapies have been defined for use after treatment failure of these agents. A phase II trial was thus conducted with paclitaxel in patients with advanced KS to assess safety and antitumor activity. MATERIALS AND METHODS: A regimen of paclitaxel at a dose of 100 mg/m(2) was given every 2 weeks to patients with advanced AIDS-related KS. Patients were treated until complete remission, disease progression, or unacceptable toxicity occurred. RESULTS: Fifty-six patients with advanced AIDS-related KS were accrued. Tumor-associated edema was present in 70% of patients and visceral involvement in 45%. Forty patients (71%) had received prior systemic therapy; 31 of these were resistant to an anthracycline. The median entry CD4(+) lymphocyte count was 20 cells/mm(3) (range, 0 to 358). A median of 10 cycles (range, 1 to 54+) of paclitaxel was administered. Fifty-nine percent of patients showed complete (n = 1) or partial response (n = 32) to paclitaxel. The median duration of response was 10.4 months (range, 2.8 to 26.7+ months) and the median survival was 15.4 months. The main side effects of therapy were grade 3 or 4 neutropenia in 61% of patients and mild-to-moderate alopecia in 87%. CONCLUSION: Paclitaxel at 100 mg/m(2) given every 2 weeks is active and well tolerated in the treatment of advanced and previously treated AIDS-related KS. The median duration of response is among the longest observed for any regimen or single agent reported for AIDS-related KS. Paclitaxel at this dosage and schedule is a treatment option for patients with advanced AIDS-related KS, including those who have experienced treatment failure of prior systemic therapy.


Subject(s)
Acquired Immunodeficiency Syndrome/complications , Antineoplastic Agents, Phytogenic/therapeutic use , Paclitaxel/therapeutic use , Sarcoma, Kaposi/drug therapy , Skin Neoplasms/drug therapy , Adult , Antineoplastic Agents, Phytogenic/adverse effects , Dose-Response Relationship, Drug , Edema/diagnosis , Edema/drug therapy , Female , Gastrointestinal Neoplasms/complications , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/mortality , Humans , Lung Neoplasms/complications , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Male , Middle Aged , Paclitaxel/adverse effects , Remission Induction , Sarcoma, Kaposi/complications , Sarcoma, Kaposi/diagnosis , Sarcoma, Kaposi/mortality , Skin Neoplasms/complications , Skin Neoplasms/diagnosis , Skin Neoplasms/mortality , Survival Rate , Treatment Outcome
4.
Cancer Chemother Pharmacol ; 43 Suppl: S61-8, 1999.
Article in English | MEDLINE | ID: mdl-10357561

ABSTRACT

The positive impact on survival of traditional chemotherapeutic agents has renewed interest in developing newer cytotoxic agents and orally active compounds with improved therapeutic indices. In addition, new insights into the pathways of human tumorigenesis have led to novel approaches aimed at specific mechanism-based targets. The taxane class, of which paclitaxel was the first member, has the unique ability to promote and stabilize microtubule function directly, thereby inhibiting mitotic progression and inducing apoptotic cell death. Paclitaxel provides treatment benefit in a broad range of solid tumors including breast, ovarian, and lung cancer. The success with paclitaxel stimulated interest in the microtubule as a new therapeutic target. Taxane analogues with improved preclinical efficacy have been identified and are entering clinical trials. The enthusiasm for oral anticancer agents and the therapeutic importance of platinum compounds has led to the development of JM216 (satraplatin), a novel platinum IV coordination complex with oral activity in cisplatin-resistant cell lines, which is now in phase III trials in prostate cancer. Another compound in late development is DPPE, a chemopotentiator that enhances the in vivo antitumor effects of cytotoxic agents such as doxorubicin, cyclophosphamide, and cisplatin. Agents that inhibit topoisomerase I and II have also been of interest. TAS-103 is a dual topoisomerase I and II inhibitor with preclinical efficacy in a broad spectrum of tumors and in multidrug-resistant tumor cell lines. Vaccination strategies represent a rational therapeutic approach in the minimal residual disease or high-risk adjuvant therapy setting. The GMK and MGV vaccines utilizing ganglioside antigens overexpressed on human tumors such as melanoma and small cell lung cancer appear to induce antibody production reliably at tolerable doses and are under further clinical investigation. Inhibition of matrix metalloproteinases (MMPs) is another attractive target for intervention in several aspects of tumor progression. Local production of MMPs with subsequent degradation of the extracellular matrix is implicated in supporting tumor growth, invasion, and angiogenesis. The development of orally active, nontoxic MMP inhibitors is critical since these compounds will likely require chronic administration in conjunction with other therapies. Oncogenes and tumor suppressor genes are appealing targets for therapy since they are thought to be responsible for a significant number of cancers. Mutations in the Ras oncogene occur with great frequency in a number of human cancers including lung, pancreas, and colon cancer. Clinical development of potent and selective inhibitors of farnesyltransferase, the Ras-processing enzyme, is ongoing. These compounds uncouple Ras activity, affect tumor growth, and have demonstrated significant antitumor activity against experimental models of human cancer. The exciting compounds and novel therapeutic approaches currently under investigation by Bristol-Myers Squibb Pharmaceutical Research Institute offer great potential as effective cancer chemotherapy agents for the near future.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Animals , Clinical Trials as Topic , Humans
5.
Br J Cancer ; 77(11): 2020-7, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9667686

ABSTRACT

BMS-181174 is an aminodisulphide derivative of Mitomycin C (MMC) with activity against a range of tumour cell lines and xenografts, including MMC-resistant tumours. In a phase I study of 82 patients with confirmed malignancy, we administered BMS-181174 at doses of 0.8-75 mg m(-2) by intravenous injection every 28 days. At least three patients were evaluated at each dose level, and 174 courses were administered. The pharmacokinetics were dose linear at BMS-181174 doses of 11.5-75 mg m(-2) and the drug appeared to undergo wide distribution. The maximum-tolerated dose was 65 mg m(-2) in previously treated patients and 75 mg m(-2) in chemotherapy-naive cases. The dose-limiting toxicity was myelosuppression, particularly thrombocytopenia, which was prolonged and cumulative. Three patients treated at 65-75 mg m(-2) died suddenly with evidence of pneumonia/pneumonitis, thought to be drug-related. Other toxicities included thrombophlebitis, possible cardiotoxicity (asymptomatic, reversible decline in left ventricular function) and renal impairment. The partial response rate was 5% (4 out of 82) overall, and 9% (3 out of 32) in patients treated at 65-75 mg m(-2). Responses occurred in treated and previously-untreated patients, including cases of colorectal cancer, non-small-cell lung cancer, ovarian cancer and adenocarcinoma of unknown primary site. BMS-181174 has anti-cancer activity but, because of its toxicity, particularly pneumonitis and thrombophlebitis, no phase II studies are planned.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Mitomycins , Neoplasms/drug therapy , Adult , Aged , Female , Heart/drug effects , Humans , Kidney/drug effects , Lung/drug effects , Male , Middle Aged , Mitomycin/adverse effects , Mitomycin/pharmacokinetics , Mitomycin/therapeutic use , Thrombophlebitis/chemically induced
6.
Semin Oncol ; 24(1 Suppl 2): S2-31-S2-33, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9045333

ABSTRACT

Paclitaxel (Taxol; Bristol-Myers Squibb Company, Princeton, NJ) combined with cisplatin seems the new standard of care for ovarian cancer patients. Since carboplatin lacks the neurotoxicity of cisplatin with an equal antitumor activity against ovarian cancer, it was chosen as the next logical step for combination chemotherapy with paclitaxel. In 46 patients an alternating dose-escalation trial has been performed. The maximum tolerated doses are carboplatin 500 mg (area under the concentration-time curve of 9) and paclitaxel 200 mg/m2 given every 3 weeks. The dose-limiting toxicity is thrombocytopenia, which emerges in the later stages of the treatment. A true platelet-sparing effect of the combination seems highly probable. The antitumor activity of the combination equals that reported for the new standard paclitaxel/cisplatin treatment. Further phase III studies are warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Ovarian Neoplasms/drug therapy , Paclitaxel/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Area Under Curve , Carboplatin/administration & dosage , Carboplatin/pharmacokinetics , Disease-Free Survival , Drug Administration Schedule , Female , Humans , Middle Aged , Paclitaxel/pharmacokinetics , Remission Induction , Thrombocytopenia/chemically induced
7.
Br J Cancer ; 75(11): 1660-6, 1997.
Article in English | MEDLINE | ID: mdl-9184183

ABSTRACT

Etoposide phosphate is a water-soluble prodrug of etoposide. The plasma pharmacokinetics of etoposide following oral administration of etoposide phosphate or oral etoposide were compared. Seventeen patients with solid tumours were enrolled to receive oral etoposide phosphate 125 mg m(-2) on days 1-5 every 3 weeks, with escalation to 175 mg m(-2) from course 3 when possible. Patients were randomized to receive oral etoposide phosphate or oral etoposide on day 1 of course 1 and the alternative compound on day 1 of course 2. Fifteen patients received two or more courses and were evaluable for pharmacokinetic comparisons. The median AUC(inf) (area under the concentration vs time curve from zero to infinity) of etoposide was 77.7 mg l(-1) h after etoposide phosphate (95% CI 61.3-100.5) and 62.0 mg l(-1) h after oral etoposide (95% CI 52.2-76.9). The difference in favour of etoposide phosphate was borderline significant: median 9.9 mg l(-1) h (95% CI 0.1-32.8 mg l(-1) h; P = 0.05). However, the inter-patient variability of etoposide AUC(inf) was not improved (coefficients of variation 42.3% and 48.4%). Etoposide phosphate was undetectable in plasma after oral administration. Toxicities of oral etoposide phosphate were not different from those known for etoposide. In conclusion, oral etoposide phosphate does not offer a clinically relevant benefit over oral etoposide.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Etoposide/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Etoposide/administration & dosage , Etoposide/adverse effects , Female , Humans , Male , Middle Aged
8.
Semin Oncol ; 23(6 Suppl 13): 34-44, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8996574

ABSTRACT

The area under the plasma concentration-time curve (AUC) following an intravenous dose of etoposide varies considerably among patients, which in part contributes to the unpredictability of toxicity and response seen in individual patients. This study evaluated the utility of therapeutic drug monitoring of etoposide in reducing the interpatient variability of etoposide steady-state concentrations during prolonged infusion. The etoposide prodrug etoposide phosphate (Etopophos; Bristol-Myers Squibb Company, Princeton, NJ) was administered by infusion using an adaptive dosing strategy. It was given in combination with carboplatin, which was dosed on an AUC basis. Patients with histologically or cytologically proven small cell lung cancer were treated with etoposide phosphate by continuous 120-hour infusion and carboplatin at a dose calculated by the Calvert formula to give an AUC of 5 mg/ mL.min. Blood samples were taken on days 2 through 5 of each treatment cycle, and high-performance liquid chromatography was used to measure the plasma etoposide concentration. The resultant concentrations were compared with target concentrations of 1 or 2 micrograms/mL and these data were used to calculate the rate of infusion for the following 24 hours. In the first cohort, the target etoposide concentration was 1 microgram/mL, and this was achieved (mean +/- SD = 1.05 +/- 0.24 micrograms/mL) by infusing etoposide phosphate doses of 21 to 109 mg (15 to 68 mg/m2) per day. In the second cohort, the target concentration of 2.0 micrograms/mL was achieved (mean +/- SD = 2.05 +/- 0.31 micrograms/mL) with infused etoposide phosphate doses of 69 to 193 mg (41 to 114 mg/m2) per day. This technique reduced the variation in plasma levels and resulted in predictable hematologic toxicity. Cumulative hematologic toxicity necessitated an extension of the treatment cycle from 3 to 4 weeks, however. Of six evaluable patients, two had a complete response and one had a partial response. Therapeutic drug monitoring was shown to reduce the interpatient variation in the plasma etoposide concentration by half and shows promise for individualizing treatment with combination chemotherapy. Exploiting the known relationships between the pharmacokinetics and pharmacodynamics of these two drugs by using therapeutic drug monitoring may lead to better therapeutic safety and efficacy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/administration & dosage , Carcinoma, Small Cell/drug therapy , Etoposide/analogs & derivatives , Lung Neoplasms/drug therapy , Organophosphorus Compounds/administration & dosage , Prodrugs/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Carboplatin/pharmacokinetics , Drug Monitoring , Etoposide/administration & dosage , Etoposide/pharmacokinetics , Feasibility Studies , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Organophosphorus Compounds/pharmacokinetics , Prodrugs/pharmacokinetics
9.
J Pharmacokinet Biopharm ; 24(4): 313-25, 1996 Aug.
Article in English | MEDLINE | ID: mdl-9044163

ABSTRACT

The bioequivalence of etoposide phosphate, a prodrug of etoposide, to etoposide was assessed in a randomized, crossover study in 29 patients with histologically established solid tumors that had failed conventional treatment. Cohorts of patients received one treatment course each of etoposide and etoposide phosphate which consisted of a 100 mg/m2 per day etoposide equivalent dose infused i.v. over 1 hr on a Day 1 to 5 schedule of treatment. The second course was administered 21 days later or on recovery of blood cell counts. Plasma and urine samples were collected over 24 hr on Day 1 of each course and assayed for etoposide content by a validated HPLC/UV method. Resulting data were subjected to noncompartmental pharmacokinetic analysis. Hematology profiles were obtained by collecting blood samples prior to the first course and twice a week after each course. The pharmacodynamics and pharmacokinetics of etoposide were virtually identical after the two treatments. The point estimates (90% confidence intervals) for nadir WBC, granulocytes, hemoglobin, and platelets expressed as % decrease from the baseline, and for the pharmacokinetic parameters, Cmax, and AUC0 infinity, after intravenous etoposide phosphate relative to etoposide were 100% (96%, 105%), 97% (91%, 103%), 95% (82%, 109%), 95% (84%, 106%), 107% (101%, 113%), and 113% (107%, 119%), respectively. Therefore, etoposide phosphate is bioequivalent to etoposide based on pharmacokinetic and pharmacodynamic assessments.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Etoposide/analogs & derivatives , Etoposide/pharmacokinetics , Neoplasms/metabolism , Organophosphorus Compounds/pharmacokinetics , Prodrugs/pharmacokinetics , Adult , Aged , Analysis of Variance , Antineoplastic Agents, Phytogenic/pharmacology , Cross-Over Studies , Etoposide/pharmacology , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/drug therapy , Organophosphorus Compounds/pharmacology , Prodrugs/pharmacology , Therapeutic Equivalency
10.
J Clin Oncol ; 14(7): 2020-30, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8683232

ABSTRACT

PURPOSE: The purpose of this study was to determine the bioavailability (F) of etoposide (E;VP-16) after oral administration of the water-soluble prodrug etoposide phosphate (EP;BMY-40481) during a phase I trial in cancer patients. PATIENTS AND METHODS: Twenty-nine patients received oral EP (capsules, 50 to 150 mg/m2/d of E equivalent) for 5 days in week 1 (course 1), followed every 3 weeks thereafter by a daily intravenous (i.v.) infusion for 5 days of E (80 mg/m2, 1-hour i.v. infusion; course 2); in three patients, the i.v. E course was given before oral EP. Plasma and urine E pharmacokinetics (high-performance liquid chromatography [HPLC]) were performed on the first day of oral EP administration and on the first day of i.v. E. RESULTS: Twenty-six of 29 patients completed two courses or more, whereas three patients received only one course due to toxicity. Myelosuppression was dose-dependent and dose-limiting, with grade 4 leukoneutropenia in four of 15 patients at 125 mg/m2 and in five of seven patients at 150 mg/m2. One patient died of meningeal hemorrhage related to grade 4 thrombocytopenia. Other toxicities were infrequent and/or manageable. No objective response was observed. The maximum-tolerated dose (MTD) is therefore 150 mg/m2, and the recommended oral dose of EP for phase II trials in this poor-risk patient population is 125 mg/m2. Twenty-six patients had pharmacokinetic data for both oral EP and i.v. E, whereas three had pharmacokinetic data on the i.v. E course only. After oral administration of EP, the pharmacokinetics of E were as follows: mean absorption rate constant (Ka), 1.7 +/- 1.7 h-1 (mean +/- SD); lag time, 0.3 +/- 0.2 hours; time of maximum concentration (t(max)), 1.6 +/- 0.8 hours; and mean half-lives (t1/2), 1.6 +/- 0.2 (first) and 10.3 +/- 5.8 hours (terminal); the increase in the area under the plasma concentration-versus-time curve (AUC) of E was proportional to the EP dose. After the 1-hour i.v. infusion of E, maximum concentration (C(max)) was 15 +/- 3 micrograms/mL; mean AUC, 88.0 +/- 22.0 micrograms.h/mL; mean total-body clearance (CL), 0.97 +/- 0.24 L/h/m2 (16.2 mL/min/m2); and mean t1/2, 0.9 +/- 0.6 (first) and 8.1 +/- 4.1 hours (terminal). The 24-hour urinary excretion of E after i.v. E was significantly higher (33%) compared with that of oral EP (17%) (P < .001). Significant correlation was observed between the neutropenia at nadir and the AUC of E after oral EP administration (r = .58, P < .01, sigmoid maximum effect [E(max)] model). The mean F of E after oral administration of EP in 26 patients was 68.0 +/- 17.9% (coefficient of variation [CV], 26.3%; F range, 35.5% to 111.8%). In this study, tumor type, as well as EP dose, did not significantly influence the F in E. There was no difference in F of E, whether oral EP was administered before or after i.v. E. Compared with literature data on oral E, the percent F in E after oral prodrug EP administration was 19% higher at either low ( < or = 100 mg/m2) or high ( > 100 mg/m2) doses. CONCLUSION: Similarly to E, the main toxicity of the prodrug EP is dose-dependent leukoneutropenia, which is dose-limiting at the oral MTD of 150 mg/m2/d for 5 days. The recommended oral dose of EP is 125 mg/m2/d for 5 days every 3 weeks in poor-risk patients. Compared with literature data, oral EP has a 19% higher F value compared with oral E either at low or high doses. This higher F in E from oral prodrug EP appears to be a pharmacologic advantage that could be of potential pharmacodynamic importance for this drug.


Subject(s)
Antineoplastic Agents/administration & dosage , Etoposide/analogs & derivatives , Etoposide/pharmacokinetics , Neoplasms/drug therapy , Organophosphorus Compounds/administration & dosage , Prodrugs/administration & dosage , Administration, Oral , Adolescent , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Biological Availability , Chromatography, High Pressure Liquid , Etoposide/administration & dosage , Etoposide/adverse effects , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/metabolism , Organophosphorus Compounds/adverse effects , Organophosphorus Compounds/pharmacokinetics , Prodrugs/adverse effects , Prodrugs/pharmacokinetics
11.
J Clin Oncol ; 14(6): 1858-67, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8656254

ABSTRACT

PURPOSE AND METHODS: The objective of this multicenter study was to compare the therapeutic index of two different doses of paclitaxel given as a 3-hour infusion in patients with metastatic breast cancer (MBC), who had failed to respond to previous chemotherapy. A total of 471 patients with MBC were randomized to receive intravenous paclitaxel at a dose of 175 or 135 mg/m2 every 3 weeks. RESULTS: Better treatment results were achieved with high-dose (HD) versus low-dose (LD) paclitaxel: overall response rate, 29% versus 22% (P = .108); complete response (CR) rate, 5% versus 2% (P = .088); median time to disease progression, 4.2 versus 3.0 months (P = .027); and median survival time, 11.7 versus 10.5 months (P = .321). Patients previously exposed or resistant to anthracyclines were as likely to respond as those without such prior exposure. Treatment was well tolerated, as documented by the number of administered treatment courses (median, six v five; range, one 17 v one to 18), the low frequency of dose reductions (14% v 7%, P = .024), and the small number of patients (n = 9 or 4% vn = 5 or 2%) who required treatment discontinuation for adverse reactions. The incidence and severity of neutropenia and peripheral neuropathy were dose-related. After quality-of-life-adjusted time-to-progression analysis, the HD arm (175 mg/m2) retained its advantage over the LD arm (135 mg/m2). CONCLUSION: The results of this trial substantiate the activity of paclitaxel in the treatment of MBC. The observed superior efficacy with a dose of 175 mg/m2 over 135 mg/m2 suggests a dose-effect relationship. The clinical activity in anthracycline-resistant patients is particularly noteworthy. Paclitaxel in breast cancer needs further evaluation in large trials that use combination chemotherapy and involve earlier disease stages.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Breast Neoplasms/pathology , Paclitaxel/administration & dosage , Adult , Aged , Antineoplastic Agents, Phytogenic/adverse effects , Breast Neoplasms/mortality , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Female , Humans , Infusions, Intravenous , Middle Aged , Neoplasm Metastasis , Paclitaxel/adverse effects , Survival Rate
12.
Semin Oncol ; 22(4 Suppl 8): 16-22, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7543699

ABSTRACT

Paclitaxel (Taxol; Bristol-Myers Squibb Company, Princeton, NJ) is a novel cytostatic agent that has shown interesting antitumor activity in patients with advanced breast cancer. Depending on variable patient characteristics and amount and type of prior therapy, as well as the applied dose and schedule of paclitaxel, response rates have varied from 13% to 62%. However, optimal dose and schedule are still unknown. We studied a high-dose (250 to 300 mg/m2) 3-hour paclitaxel infusion schedule in a poor prognostic group of breast cancer patients who progressed or relapsed while taking anthracyclines. This regimen was given every 3 weeks. Twenty-one of the 36 patients studied had increased liver enzymes and 18 had documented liver metastases. The objective response rate was only 6%, but response rate by disease site indicated that soft tissue lesions responded in 30% of cases. For a better comparison with other reported data a uniform definition of "anthracycline refractory" is needed. Neuropathy, which was found to be dose limiting, and arthralgia/myalgia syndrome were the most frequently occurring toxicities. Both severe myelosuppression (and infections) and severe diarrhea and mucositis were reported more frequently in patients with liver dysfunction. As higher peak levels, increased areas under the concentration time curves, and longer times during which plasma concentrations were above the threshold level of 0.1 mumol/L were found in patients with elevated liver enzymes, a correlation with the observed toxicities is assumed. Further pharmacodynamic studies in such patients receiving a 3-hour infusion seem warranted.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Breast Neoplasms/drug therapy , Granulocyte Colony-Stimulating Factor/therapeutic use , Paclitaxel/therapeutic use , Adult , Aged , Cohort Studies , Disease Progression , Drug Resistance , Female , Humans , Infusions, Intravenous , Leukopenia/chemically induced , Liver Neoplasms/secondary , Middle Aged , Netherlands , Neutropenia/chemically induced , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Paclitaxel/pharmacokinetics , Peripheral Nervous System Diseases/chemically induced , Remission Induction , Safety , Thrombocytopenia/chemically induced
13.
Semin Oncol ; 22(3 Suppl 6): 97-100, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7597439

ABSTRACT

Second-line treatment with paclitaxel (Taxol; Bristol-Myers Squibb Company, Princeton, NJ) may achieve remissions in patients suffering from ovarian cancer who have failed primary chemotherapy with cisplatin- or carboplatin-based regimens. Introduction of paclitaxel in combination with cisplatin into the first-line treatment strategy was therefore the next logical step in the development of chemotherapy against ovarian cancer. Data already have shown that this may result in better survival. Since carboplatin may replace cisplatin, the combination of paclitaxel with carboplatin seemed a further necessary step. We therefore embarked on a dose-finding study of paclitaxel and carboplatin. Fourteen patients with International Federal of Gynecology and Obstetrics stage III and IV ovarian cancer with a median age of 55.5 years entered this study of escalating doses of either carboplatin or paclitaxel. Doses of carboplatin could be escalated from 300 to 450 mg/m2 and paclitaxel could be escalated from 125 to 175 mg/m2 without dose-limiting myelosuppression. At the highest dose level reported here, only transient short-lived leukopenia was observed. Other toxicities consisted of nausea and vomiting, peripheral neurotoxicity, and arthralgia, all mild. In the first 14 patients, 10 of whom are evaluable, complete remissions were seen in two patients and partial remissions in six. This study will escalate the doses of paclitaxel and carboplatin further. This treatment is well tolerated and yields satisfactory antitumor results.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Ovarian Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carboplatin/administration & dosage , Drug Administration Schedule , Female , Humans , Middle Aged , Paclitaxel/administration & dosage , Remission Induction
14.
Eur J Gynaecol Oncol ; 16(4): 263-7, 1995.
Article in English | MEDLINE | ID: mdl-7556281

ABSTRACT

We present a case report of a 56 year old female suffering from recurrence of simultaneous endometrial and ovarian carcinomas, confined to distant neck and axillary lymph nodes. Resistance of the tumor to conventional chemotherapy and radiotherapy was followed by experimental Paclitaxel treatment which led to complete regression of all metastatic foci.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Endometrioid/drug therapy , Carcinoma, Endometrioid/secondary , Endometrial Neoplasms/drug therapy , Neoplasms, Multiple Primary/drug therapy , Ovarian Neoplasms/drug therapy , Paclitaxel/therapeutic use , Endometrial Neoplasms/radiotherapy , Female , Humans , Lymphatic Metastasis , Middle Aged , Ovarian Neoplasms/radiotherapy
15.
Isr J Med Sci ; 30(1): 70-8, 1994 Jan.
Article in English | MEDLINE | ID: mdl-7908013

ABSTRACT

Taxol is a novel taxane derivative obtained from the bark of the Pacific yew, Taxus brevifolia, which has demonstrated substantial antitumor activity in early clinical trials. Intensive research efforts were necessary to overcome both supply problems and hypersensitivity reactions to the drug and thus assure its widespread use. Taxol is active in a variety of neoplasias, including advanced breast and ovarian tumors resistant to drugs such as doxorubicin and cisplatin, respectively. We report here the initial experience with taxol in these two disease entities in Israel, at three institutions within the framework of large multinational trials. These studies compared a) the use of two dose levels of taxol, and b) short, 3-h administration vs. a longer 24-h infusion of the drug. A total of 107 Israeli patients, 38 with ovarian cancer and 69 with breast cancer, were given 706 courses of taxol. Our results show that the administration of taxol at doses ranging between 135 and 175 mg/m2 is indeed feasible and that 3-h infusions are as well tolerated as longer administration. The main hematological toxicity was leukopenia, which was promptly reversible and was more pronounced both at the higher dose level and with the more prolonged infusion. Of the nonhematological side effects, the most prominent were alopecia, mild nausea and vomiting, limb paresthesias, fatigue and myalgia. Allergic reactions following routine premedication were mild and infrequent, never necessitating discontinuation of the drug. Clinically significant cardiac events did not occur. Taxol is an important addition to the anticancer chemotherapy armamentarium.


Subject(s)
Breast Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/drug therapy , Paclitaxel/therapeutic use , Adult , Aged , Breast Neoplasms/pathology , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Israel , Leukopenia/chemically induced , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Ovarian Neoplasms/pathology , Remission Induction , Time Factors
16.
Eur J Cancer ; 29A(11): 1566-73, 1993.
Article in English | MEDLINE | ID: mdl-8217364

ABSTRACT

The cytotoxic effects of conventional (doxorubicin, 5-fluorouracil, cisplatin) and investigational (2',2'-difluorodeoxycytidine, hexadecylphosphocholine, EO9, rhizoxin) anticancer drugs were studied in subconfluent and multilayered postconfluent cultures of human colon and ovarian carcinoma cell lines. Chemosensitivity was assessed 4 days after a 24-h drug exposure with the sulphorhodamine B assay. Except for rhizoxin, all drugs tested yielded an EC50 (drug concentration producing absorbance readings 50% lower than those of non-treated wells) in postconfluent cultures that were higher than an EC50 obtained with subconfluent cultures. Compared with subconfluent cultures, postconfluent cultures showed decreased cellular nucleotide concentrations and ATP/ADP ratios, in addition to an increased percentage of G0/G1 cells. The activity of DT-diaphorase, a reductase involved in the bioactivation of EO9, was similar in sub- and postconfluent cultures. These results indicate similarity of the postconfluent model presented with those obtained with in vivo models and more complex in vitro techniques.


Subject(s)
Antineoplastic Agents/pharmacology , Tumor Cells, Cultured/drug effects , Cell Cycle , Cell Division/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Humans , NAD(P)H Dehydrogenase (Quinone)/metabolism
17.
J Natl Cancer Inst Monogr ; (15): 131-9, 1993.
Article in English | MEDLINE | ID: mdl-7912519

ABSTRACT

The safety profile of Taxol administered intravenously as a single agent has been established based on the experience of 655 patients. Of these patients, 253 were treated in nine phase I studies, and 402 were treated in eight disease-oriented phase II studies. Myelosuppression, specifically neutropenia, was the dose-limiting toxicity in all studies conducted in patients with solid tumors. Neutropenia was schedule dependent and was less severe when Taxol was administered via a 3-hour infusion. Severe hypersensitivity reactions were controlled in the phase II program with a premedication regimen consisting of dexamethasone, an antihistamine, and an H2 blocker. Cardiovascular toxicities were minimal and do not indicate constant electrocardiographic monitoring during Taxol infusions. Peripheral neuropathy was usually mild to moderate and dose related; however, it rarely caused treatment discontinuation. Additional adverse events associated with Taxol include arthralgia/myalgia, mucositis, nausea and vomiting, and alopecia.


Subject(s)
Paclitaxel/adverse effects , Bone Marrow/drug effects , Cardiovascular Diseases/chemically induced , Drug Hypersensitivity , Humans , Nervous System Diseases/chemically induced
18.
Semin Oncol ; 19(6): 613-21, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1462163

ABSTRACT

The discovery and development of new anticancer drugs is a complex and largely empirical process. New compounds can be discovered by screening, modification of existing compounds, rational drug design, and serendipitous basic research observations. Selection of compounds for clinical trials depends on assays of uncertain predictive value. In the pharmaceutical industry, priorities for development of potentially active entities are set and available resources allocated based on the availability and cost of supplies, patent status, potential spectrum of activity, ability to meet regulatory requirements, and market assessments. Competition for resources also occurs from noncancer drugs, eg, cardiovascular agents. Clinical development (testing and approval for commercial distribution) requires close attention to the requirements of national regulatory agencies such as the United States Food and Drug Administration. The arbitrary nature by which compounds with antitumor potential are chosen for development means that some that would be useful never reach clinical trial and others are never made generally available. This article reviews the decision making process in the pharmaceutical industry by which compounds are identified and selected for clinical trial, the regulations in the United States that govern such trials, and what is required to have the drug approved for commercial distribution.


Subject(s)
Drug Industry , Drugs, Investigational , Animals , Antineoplastic Agents , Clinical Trials as Topic , Drug Approval , Drug Evaluation, Preclinical , Humans , United States
19.
Cancer Res ; 52(21): 5940-7, 1992 Nov 01.
Article in English | MEDLINE | ID: mdl-1394220

ABSTRACT

In a European joint project carried out in 6 laboratories a disease-oriented program was set up consisting of a panel of 7 tumor types, each represented by 4 to 8 different human tumor lines, for secondary screening of promising anticancer drugs. Human tumor lines were selected on the basis of differences in histology, growth rate, and sensitivity to conventional cytostatic agents. Xenografts were grown s.c. in nude mice, and treatment was started when tumors reached a mean diameter of 6 mm in groups of mice where at least 6 tumors were evaluable. Drugs were given at the maximum tolerated dose. For evaluation of drug efficacy, median tumor growth curves were drawn, and specific growth delay and treated/control x 100% were calculated. Doxorubicin (8 mg/kg i.v. days 1 and 8) was effective (treated/control < 50%, and specific growth delay > 1.0) in 0 of 2 breast cancers, 1 of 3 colorectal cancers, 2 of 5 head and neck cancers, 3 of 6 non-small cell lung cancers, 4 of 6 small cell lung cancers, 0 of 3 melanomas, and 3 of 6 ovarian cancer lines. Amsacrine (8 mg/kg i.v. days 1 and 8) was not effective, while datelliptium (35 mg/kg i.p. days 1 and 8) was active against 2 of 6 small cell lung cancer lines. Brequinar sodium (50 mg/kg i.p. days 1-5) showed efficacy in 4 of 5 head and neck cancers, 5 of 8 non-small cell lung cancers, and 4 of 5 small cell lung cancer lines. The project has been shown to be a feasible approach. Clinical activity for doxorubicin and inactivity for amsacrine against solid tumor types was confirmed in the human tumor xenograft panel. Additional anticancer drugs will be studied in the European joint project to further define the reliability of this novel, promising screening approach.


Subject(s)
Amsacrine/pharmacology , Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Doxorubicin/pharmacology , Ellipticines/pharmacology , Animals , Body Weight/drug effects , Drug Screening Assays, Antitumor , Europe , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Transplantation, Heterologous , Tumor Cells, Cultured
20.
Br J Cancer ; 66(4): 660-5, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1419603

ABSTRACT

This study assessed the growth pattern, cellular organisation and chemosensitivity of established human tumour cell lines growing as postconfluent cultures in 'V'-bottomed, 96-well microtiter plates. Cross-sections of the colon (HT29, SW620, SW1116), ovarian (A2780) and head and neck (UM-SCC-22B) carcinoma microcultures allowed in situ evaluation of the cellular organisation in the wells. After 5 days of growth, every cell line had reached confluence, but each of them displayed a specific pattern of cell stacking which ranged from two to ten layers. Postconfluent HT29 cells displayed morphologic features suggestive of some degree of enterocytic differentiation. Growth and cytotoxicity could be studied reliably and reproducibly in this system with the sulforhodamine B protein assay. Against HT29 postconfluent cultures, the EC50's (drug concentrations producing absorbance readings 50% lower than those of non-treated wells) of 5-fluorouracil and of the ether lipid, hexadecylphosphocholine, were 1 mM and 50 microM respectively. The possibility to perform chemosensitivity tests using semiautomated microtiter plate technology supports further evaluation of this system as an alternative antitumour drug testing model.


Subject(s)
Drug Screening Assays, Antitumor/methods , Tumor Cells, Cultured/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Division , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Feasibility Studies , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Rhodamines
SELECTION OF CITATIONS
SEARCH DETAIL
...