Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 29
Filtrer
1.
J Clin Oncol ; 41(36): 5482-5492, 2023 12 20.
Article de Anglais | MEDLINE | ID: mdl-38100992

RÉSUMÉ

PURPOSE: Most patients with advanced pancreas cancer experience pain and must limit their daily activities because of tumor-related symptoms. To date, no treatment has had a significant impact on the disease. In early studies with gemcitabine, patients with pancreas cancer experienced an improvement in disease-related symptoms. Based on those findings, a definitive trial was performed to assess the effectiveness of gemcitabine in patients with newly diagnosed advanced pancreas cancer. PATIENTS AND METHODS: One hundred twenty-six patients with advanced symptomatic pancreas cancer completed a lead-in period to characterize and stabilize pain and were randomized to receive either gemcitabine 1,000 mg/m2 weekly x 7 followed by 1 week of rest, then weekly x 3 every 4 weeks thereafter (63 patients), or to fluorouracil (5-FU) 600 mg/m2 once weekly (63 patients). The primary efficacy measure was clinical benefit response, which was a composite of measurements of pain (analgesic consumption and pain intensity), Karnofsky performance status, and weight. Clinical benefit required a sustained (> or = 4 weeks) improvement in at least one parameter without worsening in any others. Other measures of efficacy included response rate, time to progressive disease, and survival. RESULTS: Clinical benefit response was experienced by 23.8% of gemcitabine-treated patients compared with 4.8% of 5-FU-treated patients (P = .0022). The median survival durations were 5.65 and 4.41 months for gemcitabine-treated and 5-FU-treated patients, respectively (P = .0025). The survival rate at 12 months was 18% for gemcitabine patients and 2% for 5-FU patients. Treatment was well tolerated. CONCLUSION: This study demonstrates that gemcitabine is more effective than 5-FU in alleviation of some disease-related symptoms in patients with advanced, symptomatic pancreas cancer. Gemcitabine also confers a modest survival advantage over treatment with 5-FU.

2.
Article de Anglais | MEDLINE | ID: mdl-27464353

RÉSUMÉ

The symptom literature in cancer has primarily examined symptom severity, frequency and distress. Assessing cancer patients' perceptions of symptom importance-how important it is for them to see improvement in a symptom following an intervention-and factors influencing these judgments would also inform patient-centred care, but this analysis has not been undertaken. This qualitative study aimed to identify factors underlying perceptions of symptom importance among 25 symptomatic metastatic breast cancer (MBC) patients. Participants were recruited from a cancer centre in the Midwestern USA. Semi-structured interviews focused on patients' rationale for considering common symptoms (i.e., anxiety, sadness, sleep problems, pain or fatigue) to be important. Thematic analyses revealed five interrelated factors underlying MBC patients' perceptions of symptom importance: activity restriction, concentration difficulties, exacerbation of other physical symptoms, symptom-related long-term health concerns and negative impact on their relationships with others. Patients most frequently stated that a physical or psychological symptom was important because of the resulting activity restriction. Additionally, some patients considered pain to be important because it signalled potential long-term health concerns, such as worsening metastatic disease. Findings suggest that clinicians should take into account MBC patients' perceptions of symptom importance and factors underlying these judgments when making shared treatment decisions.


Sujet(s)
Attitude envers la santé , Tumeurs du sein/psychologie , Activités de la vie quotidienne , Adulte , Sujet âgé , Anxiété/étiologie , Attention/physiologie , Émotions , Fatigue/étiologie , Femelle , Humains , Relations interpersonnelles , Adulte d'âge moyen , États du Centre-Ouest des États-Unis , Métastase tumorale , Douleur/psychologie , Perception
3.
Pharmacogenomics J ; 17(6): 521-527, 2017 12.
Article de Anglais | MEDLINE | ID: mdl-27549341

RÉSUMÉ

Discovery of clinical and genetic predictors of exemestane pharmacokinetics was attempted in 246 postmenopausal patients with breast cancer enrolled on a prospective clinical study. A sample was collected 2 h after exemestane dosing at a 1- or 3-month study visit to measure drug concentration. The primary hypothesis was that patients carrying the low-activity CYP3A4*22 (rs35599367) single-nucleotide polymorphism (SNP) would have greater exemestane concentration. Additional SNPs in genes relevant to exemestane metabolism (CYP1A1/2, CYP1B1, CYP3A4, CYP4A11, AKR1C3/4, AKR7A2) were screened in secondary analyses and adjusted for clinical covariates. CYP3A4*22 was associated with a 54% greater exemestane concentration (P<0.01). Concentration was greater in patients who reported White race, had elevated aminotransferases, renal insufficiency, lower body mass index and had not received chemotherapy (all P<0.05), and CYP3A4*22 maintained significance after adjustment for covariates (P<0.01). These genetic and clinical predictors of exemestane concentration may be useful for treatment individualization in patients with breast cancer.


Sujet(s)
Androstadiènes/sang , Antinéoplasiques/sang , Tumeurs du sein/génétique , Cytochrome P-450 CYP3A/génétique , Variants pharmacogénomiques , Polymorphisme de nucléotide simple , Androstadiènes/administration et posologie , Androstadiènes/usage thérapeutique , Antinéoplasiques/administration et posologie , Antinéoplasiques/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/enzymologie , Femelle , Techniques de génotypage , Humains , Adulte d'âge moyen , Test pharmacogénomique , Post-ménopause , Médecine de précision , Valeur prédictive des tests
4.
Br J Cancer ; 109(9): 2331-9, 2013 Oct 29.
Article de Anglais | MEDLINE | ID: mdl-24084768

RÉSUMÉ

BACKGROUND: Change in breast density may predict outcome of women receiving adjuvant hormone therapy for breast cancer. We performed a prospective clinical trial to evaluate the impact of inherited variants in genes involved in oestrogen metabolism and signalling on change in mammographic percent density (MPD) with aromatase inhibitor (AI) therapy. METHODS: Postmenopausal women with breast cancer who were initiating adjuvant AI therapy were enrolled onto a multicentre, randomised clinical trial of exemestane vs letrozole, designed to identify associations between AI-induced change in MPD and single-nucleotide polymorphisms in candidate genes. Subjects underwent unilateral craniocaudal mammography before and following 24 months of treatment. RESULTS: Of the 503 enrolled subjects, 259 had both paired mammograms at baseline and following 24 months of treatment and evaluable DNA. We observed a statistically significant decrease in mean MPD from 17.1 to 15.1% (P<0.001), more pronounced in women with baseline MPD ≥20%. No AI-specific difference in change in MPD was identified. No significant associations between change in MPD and inherited genetic variants were observed. CONCLUSION: Subjects with higher baseline MPD had a greater average decrease in MPD with AI therapy. There does not appear to be a substantial effect of inherited variants in biologically selected candidate genes.


Sujet(s)
Inhibiteurs de l'aromatase/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Région mammaire/effets des médicaments et des substances chimiques , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Androstadiènes/usage thérapeutique , Aromatase/génétique , Région mammaire/métabolisme , Région mammaire/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Traitement médicamenteux adjuvant/méthodes , Oestrogènes/métabolisme , Femelle , Humains , Létrozole , Mammographie/méthodes , Adulte d'âge moyen , Nitriles/usage thérapeutique , Polymorphisme de nucléotide simple , Post-ménopause/effets des médicaments et des substances chimiques , Post-ménopause/génétique , Post-ménopause/métabolisme , Études prospectives , Triazoles/usage thérapeutique
5.
Clin Pharmacol Ther ; 90(5): 693-700, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21975350

RÉSUMÉ

The associations between plasma letrozole concentrations and CYP2A6 and CYP3A5 genetic variants were tested in the Exemestane and Letrozole Pharmacogenomics (ELPH) trial. ELPH is a multicenter, open-label prospective clinical trial in women randomly assigned (n≈250 in each arm) to receive 2 years of treatment with either oral letrozole (2.5 mg/day) or oral exemestane (25 mg/day). CYP2A6 and CYP3A showed effects on letrozole metabolism in vitro. DNA samples were genotyped for variants in the CYP2A6 and CYP3A5 genes. Plasma letrozole concentrations showed high interpatient variability (>10-fold) and were associated significantly with CYP2A6 genotypes (P<0.0001), body mass index (BMI) (P<0.0001), and age (P=0.0035). However, CYP3A5 genotypes showed no association with plasma letrozole concentrations. These data suggest that CYP2A6 is the principal clearance mechanism for letrozole in vivo. CYP2A6 metabolic status, along with BMI and age, may serve as a biomarker of the efficacy of letrozole treatment or a predictor of adverse effects.


Sujet(s)
Antinéoplasiques/pharmacocinétique , Aryl hydrocarbon hydroxylases/génétique , Tumeurs du sein/traitement médicamenteux , Nitriles/pharmacocinétique , Post-ménopause , Triazoles/pharmacocinétique , Administration par voie orale , Adulte , Facteurs âges , Sujet âgé , Sujet âgé de 80 ans ou plus , Androstadiènes/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Indice de masse corporelle , Études croisées , Cytochrome P-450 CYP2A6 , Cytochrome P-450 CYP3A/génétique , Femelle , Variation génétique , Génotype , Humains , Létrozole , Adulte d'âge moyen , Nitriles/usage thérapeutique , Pharmacogénétique , Études prospectives , Triazoles/usage thérapeutique
6.
Climacteric ; 14(1): 171-80, 2011 Feb.
Article de Anglais | MEDLINE | ID: mdl-20450413

RÉSUMÉ

OBJECTIVES: To evaluate the relationships among measures of hot flushes, perceived hot flush interference, sleep disturbance, and measures of quality of life while controlling for potential covariates (patient and treatment variables). METHODS: Breast cancer survivors (n = 395) due to receive aromatase inhibitor therapy provided demographic information, physiological hot flush data via sternal skin conductance monitoring, hot flush frequency via written diary and electronic event marker, hot flush severity and bother via written diary, and questionnaire data via the Hot Flash Related Daily Interference Scale, Pittsburgh Sleep Quality Index, the EuroQOL, Hospital Anxiety and Depression Scale and the Center for Epidemiologic Studies Depression Scale. RESULTS: Confirmatory factor analysis supported a two-factor model for hot flush symptoms (frequency and severity). Although there was strong convergence among self-reported hot flush measures, there was a high degree of unexplained variance associated with physiological measures. This suggests that self-report and physiological measures do not overlap substantially. The structural model showed that greater hot flush frequency and severity were directly related to greater perceived interference with daily life activities. Greater perceived interference, in turn, directly predicted greater sleep disruption, which predicted lower perceived health state and more symptoms of anxiety and depression. CONCLUSIONS: Findings suggest hot flush interference may be the most appropriate single measure to include in clinical trials of vasomotor symptom therapies. Measuring and ameliorating patients' perceptions of hot flush interference with life activities and subjective sleep quality may be the most direct routes to improving quality of life.


Sujet(s)
Tumeurs du sein/psychologie , Analyse statistique factorielle , Bouffées de chaleur/psychologie , Modèles biologiques , Qualité de vie , Anxiété/psychologie , Inhibiteurs de l'aromatase/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Dépression/psychologie , Femelle , Réflexe psychogalvanique , État de santé , Humains , Adulte d'âge moyen , Surveillance électronique ambulatoire , Indice de gravité de la maladie , Troubles de la veille et du sommeil/psychologie , Enquêtes et questionnaires , Survivants
7.
Br J Cancer ; 103(3): 291-6, 2010 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-20606683

RÉSUMÉ

BACKGROUND: The aromatase inhibitor (AI)-associated musculoskeletal syndrome (AIMSS) occurs in approximately 50% of AI-treated patients. Inflammatory mediators are associated with oestrogen signalling and may change with oestrogen depletion. We hypothesised that AIMSS may be associated with changes in circulating inflammatory markers. METHODS: Patients with breast cancer were enrolled in a trial of adjuvant AI therapy. Changes in pain and function during therapy were assessed prospectively. We selected 30 cases with AIMSS and 22 controls without AIMSS, matched for demographics and prior therapy. Serum samples collected at baseline and during treatment were assayed for multiple inflammatory cytokines and lipid mediators using multiplex assays. RESULTS: Before AI therapy, mean serum concentrations of 6 of 36 assayed factors were statistically significantly lower in cases than controls (all P<0.003). No statistically significant changes during AI therapy relative to pre-treatment were observed between cases and controls for any of the inflammatory markers tested. CONCLUSION: AIMSS is probably not associated with a systemic inflammatory response. Pre-treatment cytokine levels may predict for development of AIMSS.


Sujet(s)
Androstadiènes/usage thérapeutique , Inhibiteurs de l'aromatase/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Cytokines/sang , Inflammation/induit chimiquement , Maladies ostéomusculaires/induit chimiquement , Sujet âgé , Antinéoplasiques/usage thérapeutique , Inhibiteurs de l'aromatase/effets indésirables , Tumeurs du sein/sang , Composés pontés/usage thérapeutique , Études cas-témoins , Oestrogènes/déficit , Femelle , Humains , Adulte d'âge moyen , Post-ménopause , Syndrome , Tamoxifène/usage thérapeutique , Taxoïdes/usage thérapeutique
8.
Br J Cancer ; 102(2): 294-300, 2010 Jan 19.
Article de Anglais | MEDLINE | ID: mdl-19953095

RÉSUMÉ

BACKGROUND: Tamoxifen, a selective oestrogen receptor (ER) modulator, increases bone mineral density (BMD) in postmenopausal women and decreases BMD in premenopausal women. We hypothesised that inherited variants in candidate genes involved in oestrogen signalling and tamoxifen metabolism might be associated with tamoxifen effects in bone. METHODS: A total of 297 women who were initiating tamoxifen therapy were enrolled in a prospective multicentre clinical trial. Lumbar spine and total hip BMD values were measured using dual-energy X-ray absorptiometry (DXA) at baseline and after 12 months of tamoxifen therapy. Single-nucleotide polymorphisms (SNPs) in ESR1, ESR2, and CYP2D6 were tested for associations in the context of menopausal status and previous chemotherapy, with a mean percentage change in BMD over 12 months. RESULTS: The percentage increase in BMD was greater in postmenopausal women and in those patients who had been treated with chemotherapy. No significant associations between tested SNPs and either baseline BMD or change in BMD with 1 year of tamoxifen therapy were detected. CONCLUSION: The evaluated SNPs in ESR and CYP2D6 do not seem to influence BMD in tamoxifen-treated subjects.


Sujet(s)
Antinéoplasiques hormonaux/pharmacologie , Densité osseuse/effets des médicaments et des substances chimiques , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Récepteur alpha des oestrogènes/génétique , Tamoxifène/pharmacologie , Absorptiométrie photonique , Adulte , Cytochrome P-450 CYP2D6/génétique , Récepteur bêta des oestrogènes/génétique , Femelle , Humains , Adulte d'âge moyen , Polymorphisme de nucléotide simple , Études prospectives , Enregistrements
9.
Pharmacogenomics J ; 9(4): 258-64, 2009 Aug.
Article de Anglais | MEDLINE | ID: mdl-19421167

RÉSUMÉ

The selective estrogen receptor modulator tamoxifen is routinely used for treatment and prevention of estrogen-receptor-positive breast cancer. Studies of tamoxifen adherence suggest that over half of patients discontinue treatment before the recommended 5 years. We hypothesized that polymorphisms in CYP2D6, the enzyme responsible for tamoxifen activation, predict for tamoxifen discontinuation. Tamoxifen-treated women (n=297) were genotyped for CYP2D6 variants and assigned a 'score' based on predicted allele activities from 0 (no activity) to 2 (high activity). Correlation between CYP2D6 score and discontinuation rates at 4 months was tested. We observed a strong nonlinear correlation between higher CYP2D6 score and increased rates of discontinuation (r(2)=0.935, P=0.018). These data suggest that presence of active CYP2D6 alleles may predict for higher likelihood of tamoxifen discontinuation. Therefore, patients who may be most likely to benefit from tamoxifen may paradoxically be most likely to discontinue treatment prematurely.


Sujet(s)
Antinéoplasiques hormonaux/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Cytochrome P-450 CYP2D6/génétique , Observance par le patient , Modulateurs sélectifs des récepteurs des oestrogènes/usage thérapeutique , Tamoxifène/usage thérapeutique , Cytochrome P-450 CYP2D6/métabolisme , Femelle , Humains , Études prospectives , Tamoxifène/effets indésirables , Tamoxifène/métabolisme
10.
Ann Oncol ; 19(6): 1068-74, 2008 Jun.
Article de Anglais | MEDLINE | ID: mdl-18283035

RÉSUMÉ

BACKGROUND: The efficacy and tolerability of the epidermal growth factor receptor/human epidermal growth factor receptor type 2 (HER2) tyrosine kinase inhibitor lapatinib in refractory metastatic breast cancer were assessed. PATIENTS AND METHODS: In a phase II, open-label study, patients with previously treated HER2-positive (n = 140) or HER2-negative (n = 89) metastatic breast cancer received once-daily oral lapatinib 1500 mg/day. RESULTS: Most (76%) patients had received four or more lines of prior therapy. The response rate in the HER2-positive cohort was 4.3% by investigator assessment and 1.4% by independent assessment. Both assessments established that approximately 6% of HER2-positive patients derived clinical benefit from lapatinib, being progression free for >/=6 months. No objective tumor responses occurred in the HER2-negative cohort. Independent review assessments of median time to progression and median progression-free survival were similar in the HER2-positive and HER2-negative cohorts (9.1 and 7.6 weeks, respectively). All responders exhibited HER2 overexpression (3+ by immunohistochemistry), and five of six responders were HER2 amplified by FISH. Lapatinib-related adverse events, including diarrhea (54%), rash (30%), and nausea (24%), were primarily mild to moderate in severity. CONCLUSIONS: Lapatinib monotherapy had modest clinical activity in HER2-positive metastatic breast cancer that progressed on prior trastuzumab regimens. No apparent clinical activity was observed in chemotherapy-refractory, HER2-negative disease.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Gènes erbB-2 , Quinazolines/usage thérapeutique , Adulte , Sujet âgé , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/secondaire , Femelle , Humains , Lapatinib , Adulte d'âge moyen
11.
Cancer ; 85(9): 2023-32, 1999 May 01.
Article de Anglais | MEDLINE | ID: mdl-10223245

RÉSUMÉ

BACKGROUND: Hemolytic uremic syndrome (HUS) is a rare condition that occasionally is reported in cancer patients. Recently it has been observed that gemcitabine rarely may be associated with this condition. METHODS: The manufacturer's safety database and literature were reviewed for any report regarding gemcitabine associated with renal and hematologic abnormalities. Descriptive analysis was used to examine each case for an association between gemcitabine therapy and HUS and to identify its incidence and risk factors. RESULTS: Through December 31, 1997, 12 cases were identified that fit either the clinical (uremia, microangiopathic hemolytic anemia, and thrombocytopenia) or pathologic (renal biopsy) criteria for HUS. There were 7 males (58%) and 5 females (42%) with a median age of 55.5 years (range, 37-73 years). The median duration of gemcitabine therapy was 5.8 months (range, 3.8-13.1 months). Six patients died, five improved, and one patient's outcome was unknown. Among the six deaths, three patients died of cancer progression, one patient died of an unrelated myocardial infarction, and two patients died of HUS or HUS-related complications. For the five patients who improved, treatment was comprised of dialysis, plasmapheresis, splenectomy, or a combination. Attempts to correlate patient demographics, primary malignancy, and cumulative gemcitabine dose failed to identify consistent risk factors in predisposing patients to HUS. Confounding factors were common, including mitomycin-C and/or 5-fluorouracil exposure, advanced stage tumors, or preexisting renal dysfunction. CONCLUSIONS: Based on a patient exposure of 78,800, a crude overall incidence rate of 0.015% (range, 0.008-0.078%) was determined, showing that HUS associated with gemcitabine treatment appears to be rare. Nonetheless, as with other cancer treatments, clinicians should weigh the appropriate risk/benefit ratio in using gemcitabine to treat their patients.


Sujet(s)
Antimétabolites antinéoplasiques/usage thérapeutique , Désoxycytidine/analogues et dérivés , Syndrome hémolytique et urémique/traitement médicamenteux , Adulte , Sujet âgé , , Désoxycytidine/usage thérapeutique , Femelle , Syndrome hémolytique et urémique/épidémiologie , Humains , Incidence , Mâle , Adulte d'âge moyen , Études rétrospectives , Facteurs de risque , Résultat thérapeutique , États-Unis/épidémiologie ,
12.
Cancer ; 85(6): 1261-8, 1999 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-10189130

RÉSUMÉ

BACKGROUND: An Investigational New Drug (IND) treatment program allows patients access to a drug that has shown activity against a serious or life-threatening disease prior to full Food and Drug Administration (FDA) review and approval. This treatment IND program, in which patients with locally advanced or metastatic pancreatic carcinoma were treated with gemcitabine, began in 1995. METHODS: Eligibility criteria were < or =1 prior chemotherapy regimen; a Karnofsky performance status (KPS) of > or =50; and adequate bone marrow, liver, and renal function. Gemcitabine was given at a dose of 1000 mg/m2 weekly x 7 followed by a week of rest, then weekly x 3 every 4 weeks thereafter. In this program, disease-related symptom improvement (DRSI) was defined retrospectively as 1) improvement in pain (on a 7-point scale) and/or analgesic class (e.g., morphine improving to codeine) and/or KPS (> or =20 points), or 2) stability of these three parameters with a 7% increase in weight from baseline. RESULTS: A total of 3023 patients enrolled. At baseline, 80% of them had Stage IV disease, and 84% had a baseline KPS > or = 70. The median age was 65 years, and 56% of the patients were male. The cumulative DRSI response rate after the fourth cycle was 18.4%. Of 982 patients with tumor response data, there were 14 with complete response and 104 with partial response, for an overall response rate of 12.0% (95% confidence interval [CI], 10.0-14.0%). For 2380 patients with survival data, the median survival was 4.8 months (95% CI, 4.5-5.1 months) and the 12-month survival was 15%. Gemcitabine was well tolerated; only 4.6% of discontinuations were due to adverse events. CONCLUSIONS: Notable disease-related symptom improvement and survival were seen with gemcitabine in this large, compassionate-use setting, and these findings were in agreement with those of earlier registration trials.


Sujet(s)
Antimétabolites antinéoplasiques/usage thérapeutique , Carcinomes/traitement médicamenteux , Désoxycytidine/analogues et dérivés , Tumeurs du pancréas/traitement médicamenteux , Sujet âgé , Antimétabolites antinéoplasiques/effets indésirables , Carcinomes/mortalité , Désoxycytidine/effets indésirables , Désoxycytidine/usage thérapeutique , Médicaments en essais cliniques/effets indésirables , Médicaments en essais cliniques/usage thérapeutique , Femelle , Humains , Mâle , Adulte d'âge moyen , Tumeurs du pancréas/mortalité , Taux de survie ,
13.
J Clin Oncol ; 15(6): 2403-13, 1997 Jun.
Article de Anglais | MEDLINE | ID: mdl-9196156

RÉSUMÉ

PURPOSE: Most patients with advanced pancreas cancer experience pain and must limit their daily activities because of tumor-related symptoms. To date, no treatment has had a significant impact on the disease. In early studies with gemcitabine, patients with pancreas cancer experienced an improvement in disease-related symptoms. Based on those findings, a definitive trial was performed to assess the effectiveness of gemcitabine in patients with newly diagnosed advanced pancreas cancer. PATIENTS AND METHODS: One hundred twenty-six patients with advanced symptomatic pancreas cancer completed a lead-in period to characterize and stabilize pain and were randomized to receive either gemcitabine 1,000 mg/m2 weekly x 7 followed by 1 week of rest, then weekly x 3 every 4 weeks thereafter (63 patients), or to fluorouracil (5-FU) 600 mg/m2 once weekly (63 patients). The primary efficacy measure was clinical benefit response, which was a composite of measurements of pain (analgesic consumption and pain intensity), Karnofsky performance status, and weight. Clinical benefit required a sustained (> or = 4 weeks) improvement in at least one parameter without worsening in any others. Other measures of efficacy included response rate, time to progressive disease, and survival. RESULTS: Clinical benefit response was experienced by 23.8% of gemcitabine-treated patients compared with 4.8% of 5-FU-treated patients (P = .0022). The median survival durations were 5.65 and 4.41 months for gemcitabine-treated and 5-FU-treated patients, respectively (P = .0025). The survival rate at 12 months was 18% for gemcitabine patients and 2% for 5-FU patients. Treatment was well tolerated. CONCLUSION: This study demonstrates that gemcitabine is more effective than 5-FU in alleviation of some disease-related symptoms in patients with advanced, symptomatic pancreas cancer. Gemcitabine also confers a modest survival advantage over treatment with 5-FU.


Sujet(s)
Antimétabolites antinéoplasiques/usage thérapeutique , Désoxycytidine/analogues et dérivés , Fluorouracil/usage thérapeutique , Tumeurs du pancréas/traitement médicamenteux , Adulte , Sujet âgé , Antimétabolites antinéoplasiques/effets indésirables , Désoxycytidine/effets indésirables , Désoxycytidine/usage thérapeutique , Femelle , Fluorouracil/effets indésirables , Humains , Pompes à perfusion , Mâle , Adulte d'âge moyen , Morphine/usage thérapeutique , Stupéfiants/usage thérapeutique , Douleur/traitement médicamenteux , Tumeurs du pancréas/mortalité , Taux de survie , Résultat thérapeutique ,
14.
Semin Oncol ; 24(2 Suppl 7): S7-2-S7-7, 1997 Apr.
Article de Anglais | MEDLINE | ID: mdl-9194473

RÉSUMÉ

Gemcitabine (2',2'-difluorodeoxycytidine) is a novel nucleoside analogue that exerts its antitumor activity via multiple mechanisms of action. These include (1) incorporation of gemcitabine into replicating DNA, which inhibits DNA replication and cell growth, (2) masked DNA chain termination, and (3) several self-potentiation mechanisms that serve to increase intracellular levels of the active compound. Preclinical experiments in various cell lines and animal models demonstrate a broad range of cytotoxic activity. Pharmacokinetic studies of gemcitabine delivered by its usual schedule (30-minute weekly infusion) reveal a short plasma half-life and a high clearance into central and peripheral compartments (two-compartment model). The drug is excreted almost completely in the urine as the parent compound and primary metabolite (difluorodeoxyuridine). Phase I trials demonstrate that pharmacokinetics are schedule dependent and that, in general, gemcitabine is well tolerated. Dose-limiting toxicities are primarily myelosuppression, with other toxicities being rash, flu-like symptoms, and transient elevations in liver function tests.


Sujet(s)
Antimétabolites antinéoplasiques/pharmacologie , Essais cliniques de phase I comme sujet , Désoxycytidine/analogues et dérivés , Ribonucleotide reductases/antagonistes et inhibiteurs , Animaux , Antimétabolites antinéoplasiques/pharmacocinétique , Antimétabolites antinéoplasiques/usage thérapeutique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Réplication de l'ADN/effets des médicaments et des substances chimiques , ADN tumoral/effets des médicaments et des substances chimiques , Désoxycytidine/pharmacocinétique , Désoxycytidine/pharmacologie , Désoxycytidine/usage thérapeutique , Évaluation préclinique de médicament , Période , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Sécurité ,
15.
Clin Cancer Res ; 3(4): 537-43, 1997 Apr.
Article de Anglais | MEDLINE | ID: mdl-9815717

RÉSUMÉ

We performed a pilot clinical trial with safingol (L-threo-dihydrosphingosine), a protein kinase C-specific inhibitor that potentiates the effect of doxorubicin (DOX) in tumor-bearing animals. Safingol was initially administered as a 1-h infusion at escalating doses. Fourteen days later, patients received the same dose of safingol in combination with a fixed dose of DOX. The combination was repeated at 3-week intervals. Safingol dose levels ranged from 15 to 120 mg/m2. The plasma levels achieved at the final dose level were comparable to those associated with potentiation of DOX in animals. The mean Cmax and area under the curve for safingol at the 120 mg/m2 dose level were 1040 +/- 196 ng/ml and 1251 +/- 317 mg x h/ml, respectively. The mean plasma half-life for safingol was 3.97 +/- 2.51 h, the mean estimated clearance was 3140 +/- 765 ml/min, and the mean volume of distribution was of 995 +/- 421 liters. Coadministration of a fixed dose of DOX did not significantly change the pharmacokinetics of safingol, nor did increasing doses of safingol significantly affect the pharmacokinetics of DOX. Minor responses were observed in three patients with pancreatic cancer and one patient with angiosarcoma of the scalp. This pilot Phase I study indicates that the protein kinase C inhibitor safingol can be given safely with 45 mg/m2 of DOX at a dose that is potentially pharmacologically active without dose-limiting toxicity.


Sujet(s)
Antinéoplasiques/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Antienzymes/effets indésirables , Tumeurs/traitement médicamenteux , Sphingosine/analogues et dérivés , Adulte , Sujet âgé , Antinéoplasiques/administration et posologie , Antinéoplasiques/pharmacocinétique , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/pharmacocinétique , Relation dose-effet des médicaments , Doxorubicine/administration et posologie , Calendrier d'administration des médicaments , Antienzymes/administration et posologie , Antienzymes/pharmacocinétique , Femelle , Humains , Perfusions veineuses , Leucopénie/induit chimiquement , Mâle , Taux de clairance métabolique , Adulte d'âge moyen , Tumeurs/sang , Projets pilotes , Protéine kinase C/antagonistes et inhibiteurs , Analyse de régression , Sphingosine/administration et posologie , Sphingosine/effets indésirables , Sphingosine/pharmacocinétique , Thrombopénie/induit chimiquement
16.
Eur J Cancer ; 33 Suppl 1: S18-22, 1997 Jan.
Article de Anglais | MEDLINE | ID: mdl-9166095

RÉSUMÉ

An early study with gemcitabine in pancreas cancer indicated greater relief of disease-related symptoms than expected from the objective tumour response rate. A novel design was created to assess changes in symptomatology prospectively in two studies. The design focuses on typical features seen in patients with advanced pancreas cancer (pain, impaired function, weight loss) and the endpoint is 'clinical benefit response'. Traditional endpoints of objective tumour response and survival were also included. In a randomised study, the clinical benefit response rate for gemcitabine was 24% compared with 5% for 5-fluorouracil (5-FU) (P = 0.0022). The median survival was 5.65 months for gemcitabine compared with 4.41 months for 5-FU (P = 0.0025). The corresponding objective response rates were 5.4% and 0%. Disease stabilised in 39% and 19% of gemcitabine and 5-FU patients, respectively. In a second study of 5-FU-refractory patients, 27.0% of patients were clinical benefit responders. The median survival in this second study was 3.8 months; the objective response rate was 11%, and 30% of patients had stable disease. These trials show that gemcitabine improves disease-related symptoms and survival in patients with pancreas cancer.


Sujet(s)
Antimétabolites antinéoplasiques/usage thérapeutique , Désoxycytidine/analogues et dérivés , Fluorouracil/usage thérapeutique , Tumeurs du pancréas/traitement médicamenteux , Désoxycytidine/usage thérapeutique , Humains , Études prospectives , Qualité de vie , Résultat thérapeutique ,
17.
Ann Oncol ; 7(4): 347-53, 1996 Apr.
Article de Anglais | MEDLINE | ID: mdl-8805925

RÉSUMÉ

PURPOSE: To assess the effect of gemcitabine in patients with metastatic pancreas cancer that had progressed despite prior treatment with 5-FU. PATIENTS AND METHODS: Seventy-four patients were enrolled in this multicenter trial. Alleviation of cancer-related symptoms was the primary endpoint. Sixty-three patients completed a pain stabilization period and were treated with gemcitabine. Clinical Benefit Response was defined as a > or = 50% reduction in pain intensity, > or = 50% reduction in daily analgesic consumption, or > or = 20 point improvement in KPS that was sustained for > or = 4 consecutive weeks. RESULTS: Seventeen of 63 pts (27.0%) attained a Clinical Benefit Response (95% CI: 16.0%-38.0%). The median duration of Clinical Benefit Response was 14 weeks (range: 4-69 weeks). Median survival for patients treated with gemcitabine was 3.85 months (range: 0.3-18.0+ months). Therapy was generally well-tolerated with a low incidence of grade 3 or 4 toxicities. CONCLUSION: Systematic assessment of subjective outcomes can be used to evaluate the clinical impact of new therapies for pancreas cancer, a highly symptomatic disease. Our findings suggest that gemcitabine is a useful palliative agent in patients with 5-FU-refractory pancreas cancer.


Sujet(s)
Adénocarcinome/traitement médicamenteux , Antimétabolites antinéoplasiques/usage thérapeutique , Désoxycytidine/analogues et dérivés , Fluorouracil/usage thérapeutique , Tumeurs du pancréas/traitement médicamenteux , Adénocarcinome/mortalité , Adulte , Sujet âgé , Antimétabolites antinéoplasiques/effets indésirables , Poids/effets des médicaments et des substances chimiques , Désoxycytidine/effets indésirables , Désoxycytidine/usage thérapeutique , Évolution de la maladie , Femelle , Humains , Indice de performance de Karnofsky , Mâle , Adulte d'âge moyen , Mesure de la douleur , Tumeurs du pancréas/mortalité , Reprise du traitement , Taux de survie , Résultat thérapeutique ,
18.
Laryngoscope ; 106(1 Pt 1): 37-42, 1996 Jan.
Article de Anglais | MEDLINE | ID: mdl-8544625

RÉSUMÉ

There is evidence that solid tumors rapidly acquire cellular resistance to cisplatin. This resistance is usually mild to moderate and could be circumvented with higher concentrations of drug exposure if ancillary methods were available to avoid systemic cytotoxicity. The purpose of this study was to determine whether a tenfold increase in dose (decadose) would overcome cisplatin resistance. In a clinical trial, response effects of cisplatin at dose intensities ranging from 32.5 to 200 mg/m2 per week, which were delivered by highly selective intra-arterial infusions with a simultaneously administered intravenous neutralizing agent, were measured in 31 patients with squamous cell carcinoma (SCC) of the upper aerodigestive tract (UADT). The overall response rate (complete response [CR] and partial response [PR] to cisplatin therapy at dose intensity intervals of 0 to 74, 75 to 149, and 150 to 200 mg/m2 per week were 45.5%, 72.7%, and 100%, respectively. The average received dose intensities for nonresponders and responders (CR and PR) were 57.8 and 120.7 mg/m2 per week, respectively (P = .031). The results indicate that resistance to standard doses of cisplatin by SCC of the UADT, both previously untreated and recurrent, can be substantially overcome with "decadose" cisplatin therapy. Progress toward improving survival of patients with head and neck cancer, and possibly other site-specific malignancies, may be achieved by incorporating decadose cisplatin therapy into a multimodality treatment plan.


Sujet(s)
Antinéoplasiques/administration et posologie , Carcinome épidermoïde/traitement médicamenteux , Cisplatine/administration et posologie , Tumeurs de la tête et du cou/traitement médicamenteux , Adulte , Sujet âgé , Carcinome épidermoïde/mortalité , Résistance aux médicaments antinéoplasiques , Femelle , Tumeurs de la tête et du cou/mortalité , Humains , Perfusions artérielles , Mâle , Adulte d'âge moyen , Récidive tumorale locale
19.
Cancer J Sci Am ; 1(4): 252-60, 1995.
Article de Anglais | MEDLINE | ID: mdl-9166485

RÉSUMÉ

PURPOSE: To assess whether the administration of recombinant human erythropoietin (r-HuEPO) would increase the hematocrit, reduce the requirement for transfusion, and improve the quality of life in anemic cancer patients receiving myelosuppressive, cisplatin-based chemotherapy. PATIENTS AND METHODS: One hundred thirty-two anemic cancer patients receiving cyclic, cisplatin-containing, myelosuppressive chemotherapy were evaluated. Patients received either r-HuEPO (150 U/kg) or placebo, subcutaneously, three times a week for 3 months. Responses were assessed by measuring changes in hemoglobin/hematocrit, transfusion requirement, and quality of life. RESULTS: The mean hematocrit increased by 6.0 percentage points in the r-HuEPO group versus 1.3 in the placebo group. A decrease in transfusion requirement did not reach significance over all 3 months, but there was a significant reduction in the percentage of patients transfused in the second and third months (27% r-HuEPO vs. 56% placebo) and a trend toward reduction in the mean total number of units transfused (1.20 units r-HuEPO vs. 2.02 units placebo), suggesting a lag of 1 month before r-HuEPO can affect the transfusion requirement. Pretreatment serum erythropoietin levels were lower in responders than in nonresponders (73.5 IU/L and 86.3 IU/L means, respectively). However, the magnitude of this difference was not helpful in defining which patients were likely to respond. There was a significant improvement in overall quality of life between the two treatment arms in favor of the r-HuEPO-treated group. There were no significant adverse effects associated with r-HuEPO. CONCLUSIONS: r-HuEPO is safe and can cause a significant improvement in the hematocrit and quality of life of anemic cancer patients receiving myelosuppressive, cisplatin-based chemotherapy. After 1 month of r-HuEPO, there is also a reduction in transfusion requirement.


Sujet(s)
Anémie/traitement médicamenteux , Antinéoplasiques/effets indésirables , Cisplatine/effets indésirables , Érythropoïétine/usage thérapeutique , Tumeurs/traitement médicamenteux , Adolescent , Adulte , Sujet âgé , Anémie/sang , Anémie/étiologie , Transfusion sanguine , Femelle , Humains , Mâle , Adulte d'âge moyen , Tumeurs/complications , Qualité de vie , Protéines recombinantes , Résultat thérapeutique
20.
J Clin Oncol ; 12(10): 2113-20, 1994 Oct.
Article de Anglais | MEDLINE | ID: mdl-7931481

RÉSUMÉ

PURPOSE: To determine the maximum dose-intensity of cisplatin (DDP) that could be administered by selective intraarterial (IA) infusion in combination with systemic sodium thiosulfate neutralization to patients with head and neck carcinoma. PATIENTS AND METHODS: Forty-two patients (23 untreated stage III/IV, 19 recurrent) received highly selective IA DDP, rapidly delivered through microcatheters placed angiographically, to a maximum dose-intensity of 200 mg/m2/wk. Concurrently, the systemic effects of DDP were neutralized by intravenous (IV) bolus sodium thiosulfate. RESULTS: Problems related to the infusion technique occurred in eight of 140 courses, all of which were inconsequential. The rates of reversible grade I/II and grade III/IV toxicity were 14.8% and 1.1%, respectively. Dose-limiting toxicity, which consisted of severe electrolyte loss, occurred at a dose of 200 mg/m2/wk. The maximum-tolerated dose of DDP was 150 mg/m2 administered weekly for four doses. The overall and complete response rates in 38 assessable patients were 19 of 22 (86%) and nine of 22 (41%) for stage III/IV untreated tumors and 10 of 16 (62%) and four of 16 (25%) for patients with recurrent disease, respectively. CONCLUSION: This pharmacologic strategy permits the selective and rapid delivery of extremely high doses of DDP to head and neck carcinomas with minimal procedural complications, low systemic toxicity, and high tumor response rates.


Sujet(s)
Cisplatine/administration et posologie , Tumeurs de la tête et du cou/traitement médicamenteux , Adulte , Sujet âgé , Analyse de variance , Anémie/induit chimiquement , Anémie/prévention et contrôle , Cisplatine/effets indésirables , Calendrier d'administration des médicaments , Femelle , Études de suivi , Tumeurs de la tête et du cou/mortalité , Humains , Perfusions artérielles , Mâle , Adulte d'âge moyen , Neutropénie/induit chimiquement , Neutropénie/prévention et contrôle , Projets pilotes , Induction de rémission , Taux de survie , Thiosulfates/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...