Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Article in English | MEDLINE | ID: mdl-27464353

ABSTRACT

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.


Subject(s)
Attitude to Health , Breast Neoplasms/psychology , Activities of Daily Living , Adult , Aged , Anxiety/etiology , Attention/physiology , Emotions , Fatigue/etiology , Female , Humans , Interpersonal Relations , Middle Aged , Midwestern United States , Neoplasm Metastasis , Pain/psychology , Perception
2.
Pharmacogenomics J ; 17(6): 521-527, 2017 12.
Article in English | MEDLINE | ID: mdl-27549341

ABSTRACT

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.


Subject(s)
Androstadienes/blood , Antineoplastic Agents/blood , Breast Neoplasms/genetics , Cytochrome P-450 CYP3A/genetics , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Androstadienes/administration & dosage , Androstadienes/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Female , Genotyping Techniques , Humans , Middle Aged , Pharmacogenomic Testing , Postmenopause , Precision Medicine , Predictive Value of Tests
3.
Br J Cancer ; 109(9): 2331-9, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24084768

ABSTRACT

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.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast/drug effects , Adult , Aged , Aged, 80 and over , Androstadienes/therapeutic use , Aromatase/genetics , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemotherapy, Adjuvant/methods , Estrogens/metabolism , Female , Humans , Letrozole , Mammography/methods , Middle Aged , Nitriles/therapeutic use , Polymorphism, Single Nucleotide , Postmenopause/drug effects , Postmenopause/genetics , Postmenopause/metabolism , Prospective Studies , Triazoles/therapeutic use
4.
J Clin Oncol ; 41(36): 5482-5492, 2023 12 20.
Article in English | MEDLINE | ID: mdl-38100992

ABSTRACT

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.

5.
Climacteric ; 14(1): 171-80, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20450413

ABSTRACT

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.


Subject(s)
Breast Neoplasms/psychology , Factor Analysis, Statistical , Hot Flashes/psychology , Models, Biological , Quality of Life , Anxiety/psychology , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Depression/psychology , Female , Galvanic Skin Response , Health Status , Humans , Middle Aged , Monitoring, Ambulatory , Severity of Illness Index , Sleep Wake Disorders/psychology , Surveys and Questionnaires , Survivors
6.
Br J Cancer ; 102(2): 294-300, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-19953095

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Bone Density/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Tamoxifen/pharmacology , Absorptiometry, Photon , Adult , Cytochrome P-450 CYP2D6/genetics , Estrogen Receptor beta/genetics , Female , Humans , Middle Aged , Polymorphism, Single Nucleotide , Prospective Studies , Registries
7.
Br J Cancer ; 103(3): 291-6, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20606683

ABSTRACT

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.


Subject(s)
Androstadienes/therapeutic use , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Cytokines/blood , Inflammation/chemically induced , Musculoskeletal Diseases/chemically induced , Aged , Antineoplastic Agents/therapeutic use , Aromatase Inhibitors/adverse effects , Breast Neoplasms/blood , Bridged-Ring Compounds/therapeutic use , Case-Control Studies , Estrogens/deficiency , Female , Humans , Middle Aged , Postmenopause , Syndrome , Tamoxifen/therapeutic use , Taxoids/therapeutic use
8.
Pharmacogenomics J ; 9(4): 258-64, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19421167

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Cytochrome P-450 CYP2D6/genetics , Patient Compliance , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/therapeutic use , Cytochrome P-450 CYP2D6/metabolism , Female , Humans , Prospective Studies , Tamoxifen/adverse effects , Tamoxifen/metabolism
9.
Ann Oncol ; 19(6): 1068-74, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18283035

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Genes, erbB-2 , Quinazolines/therapeutic use , Adult , Aged , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/secondary , Female , Humans , Lapatinib , Middle Aged
10.
J Clin Oncol ; 12(10): 2113-20, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7931481

ABSTRACT

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.


Subject(s)
Cisplatin/administration & dosage , Head and Neck Neoplasms/drug therapy , Adult , Aged , Analysis of Variance , Anemia/chemically induced , Anemia/prevention & control , Cisplatin/adverse effects , Drug Administration Schedule , Female , Follow-Up Studies , Head and Neck Neoplasms/mortality , Humans , Infusions, Intra-Arterial , Male , Middle Aged , Neutropenia/chemically induced , Neutropenia/prevention & control , Pilot Projects , Remission Induction , Survival Rate , Thiosulfates/therapeutic use
11.
J Clin Oncol ; 10(3): 464-73, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1740685

ABSTRACT

PURPOSE: The trial was undertaken to study the effect of administering granulocyte-macrophage colony-stimulating factor (GM-CSF) with and without peripheral-blood progenitor cells (PBPC) on the hematologic and nonhematologic toxicity observed with multiple cycles of high-dose carboplatin chemotherapy. PATIENTS AND METHODS: Eighteen patients with a variety of solid tumors received a total of 40 cycles of carboplatin, 1,200 mg/m2 per cycle, administered by continuous infusion over 96 hours. All 40 courses were administered with a daily 4-hour intravenous (IV) infusion of either 5 or 10 micrograms/kg/d of recombinant human Escherichia coli-derived GM-CSF. The first 20 courses were administered without PBPC support (treatment A). Because of severe neutropenia and thrombocytopenia, the next 20 courses of therapy were administered with GM-CSF, PBPC, and oral antibiotic prophylaxis (treatment B). RESULTS: The addition of PBPC support led to a significant reduction in the duration of neutropenia (10.5 v 7.5 days; P = .027) and thrombocytopenia (12.4 v 5.2 days; P = .001), number of RBC transfusions (six v three; P = .01) and platelet transfusions (10.3 v 3.7; P = .013), number of hospital days (12.6 v 2.9; P = .01), and days of IV antibiotics (11.8 v 2.4; P = .007) per cycle. Significant increases in the weekly dose intensity (206 v 285 mg/m2/wk; P = .014) and total dose (2,287 v 3,600 mg/m2; P = .018) of carboplatin delivered were also observed with treatment B. The overall response rate in this study was 70%, with 11 of 16 assessable patients achieving either a complete (three patients) or partial (eight patients) remission. CONCLUSION: This combination of GM-CSF and PBPC infusion represents an effective method for delivering multiple cycles of high-dose carboplatin chemotherapy and may serve as a model for the administration of high-dose chemotherapy in future trials.


Subject(s)
Carboplatin/adverse effects , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Neoplasms/drug therapy , Neutropenia/prevention & control , Stem Cell Transplantation , Thrombocytopenia/prevention & control , Adult , Carboplatin/administration & dosage , Combined Modality Therapy , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Neutropenia/chemically induced , Recombinant Proteins/therapeutic use , Thrombocytopenia/chemically induced
12.
J Clin Oncol ; 15(6): 2403-13, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9196156

ABSTRACT

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.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Fluorouracil/therapeutic use , Pancreatic Neoplasms/drug therapy , Adult , Aged , Antimetabolites, Antineoplastic/adverse effects , Deoxycytidine/adverse effects , Deoxycytidine/therapeutic use , Female , Fluorouracil/adverse effects , Humans , Infusion Pumps , Male , Middle Aged , Morphine/therapeutic use , Narcotics/therapeutic use , Pain/drug therapy , Pancreatic Neoplasms/mortality , Survival Rate , Treatment Outcome , Gemcitabine
13.
Leukemia ; 4(11): 766-70, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2232890

ABSTRACT

Thirty adult patients with CMML were evaluated to determine prognostic factors that might have an impact on conversion to acute leukemia and survival. Neither leukocyte count nor monocyte count correlated with survival. The median survival for all 30 cases was 41 months. Patients with less than 5% marrow blasts had a median survival of 60 months but those with 5-20% blasts had only a 9-month median survival.


Subject(s)
Leukemia, Myelomonocytic, Chronic/pathology , Aged , Aged, 80 and over , Bone Marrow/pathology , Chi-Square Distribution , Female , Humans , Leukemia, Myelomonocytic, Chronic/blood , Leukemia, Myelomonocytic, Chronic/mortality , Leukocyte Count , Male , Middle Aged , Monocytes , Prognosis , Regression Analysis , Retrospective Studies , Survival Rate
14.
Clin Cancer Res ; 3(4): 537-43, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9815717

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Enzyme Inhibitors/adverse effects , Neoplasms/drug therapy , Sphingosine/analogs & derivatives , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Infusions, Intravenous , Leukopenia/chemically induced , Male , Metabolic Clearance Rate , Middle Aged , Neoplasms/blood , Pilot Projects , Protein Kinase C/antagonists & inhibitors , Regression Analysis , Sphingosine/administration & dosage , Sphingosine/adverse effects , Sphingosine/pharmacokinetics , Thrombocytopenia/chemically induced
15.
Cancer Treat Rev ; 19 Suppl C: 11-20, 1993.
Article in English | MEDLINE | ID: mdl-8106154

ABSTRACT

High-dose chemotherapy regimens can cure a number of otherwise incurable diseases, such as Hodgkin's disease, non-Hodgkin's lymphoma, neuroblastoma, acute leukemia (in remission), and breast cancer. Trials of high-dose chemotherapy have generally used autologous bone marrow transplant or peripheral blood stem cell support to ensure hematologic recovery after intensive chemotherapy and/or radiation. This report describes an approach in which high-dose carboplatin chemotherapy was followed initially by granulocyte-macrophage colony-stimulating factor (GM-CSF; Escherichia coli. Sandoz-Schering, East Hanover and Kenilworth, NJ) and in subsequent patients, by both GM-CSF and repeated cycles of peripheral blood progenitor cell (PBPC) collection and administration. The addition of PBPC to this regimen led to significant reductions in the duration of neutropenia and thrombocytopenia, the requirement for erythrocyte and platelet transfusions, the length of hospital stay, and the use of intravenous antibiotics in this group relative to those patients who received GM-CSF alone. In addition, laboratory studies are presented that show a direct correlation between the number of progenitor cells reinfused and the duration of neutropenia and thrombocytopenia. The report also reviews data indicating that circulating progenitor cells are depleted by this approach. This suggests that the number of progenitor cells available for mobilization is finite. Finally, the magnitude of these effects, and their implications for future trials with repetitive cycles of dose-intensive therapy, are discussed.


Subject(s)
Carboplatin/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Transplantation , Neoplasms/therapy , Adult , Aged , Bone Marrow/drug effects , Carboplatin/adverse effects , Combined Modality Therapy , Humans , Middle Aged
16.
Eur J Cancer ; 33 Suppl 1: S18-22, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9166095

ABSTRACT

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.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Fluorouracil/therapeutic use , Pancreatic Neoplasms/drug therapy , Deoxycytidine/therapeutic use , Humans , Prospective Studies , Quality of Life , Treatment Outcome , Gemcitabine
17.
Semin Oncol ; 24(2 Suppl 7): S7-2-S7-7, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9194473

ABSTRACT

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.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Clinical Trials, Phase I as Topic , Deoxycytidine/analogs & derivatives , Ribonucleotide Reductases/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , DNA Replication/drug effects , DNA, Neoplasm/drug effects , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Evaluation, Preclinical , Half-Life , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Safety , Gemcitabine
18.
Semin Oncol ; 19(1 Suppl 2): 139-44, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1411625

ABSTRACT

Twenty patients have received 21 courses of high-dose cyclophosphamide (6 g/m2 over 4 days), etoposide (1,800 mg/m2 over 3 days), and carboplatin (800 to 1,600 mg/m2 by continuous infusion over 96 hours) and autologous bone marrow or peripheral blood stem cell rescue. The maximum tolerated dose of this regimen included these doses of cyclophosphamide and etoposide with a total of 1,600 mg/m2 carboplatin. Acute renal failure was the dose-limiting toxicity and, at the maximum tolerated dose, was observed in two patients of 14 evaluable courses. Nonhematologic toxicity was otherwise modest, and the overall response rate was 70% in patients with a wide variety of solid hematologic neoplasms.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Component Transfusion , Bone Marrow Transplantation , Carboplatin/administration & dosage , Cyclophosphamide/administration & dosage , Etoposide/administration & dosage , Acute Kidney Injury/chemically induced , Adult , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Candidiasis/etiology , Carboplatin/adverse effects , Clinical Trials, Phase I as Topic , Cyclophosphamide/adverse effects , Etoposide/adverse effects , Female , Humans , Infusions, Intravenous , Male , Neutropenia/chemically induced , Remission Induction , Stem Cells/pathology , Survival Rate , Thrombocytopenia/chemically induced
19.
Laryngoscope ; 106(1 Pt 1): 37-42, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8544625

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Cisplatin/administration & dosage , Head and Neck Neoplasms/drug therapy , Adult , Aged , Carcinoma, Squamous Cell/mortality , Drug Resistance, Neoplasm , Female , Head and Neck Neoplasms/mortality , Humans , Infusions, Intra-Arterial , Male , Middle Aged , Neoplasm Recurrence, Local
20.
Arch Otolaryngol Head Neck Surg ; 120(3): 288-92, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8123238

ABSTRACT

OBJECTIVE: Sponge-gel-supported histoculture drug-response assay (SSHDRA) represents a promising method to determine chemosensitivity of solid tumors. To determine whether the assay correlates clinically, we compared the in vivo and in vitro effects of cisplatin in 23 of 26 patients with head and neck cancers. DESIGN: The criterion for in vitro sensitivity to cisplatin was an 84% or greater inhibition by cisplatin of the number of tritiated thymidine-incorporating cells of the histocultured tumors compared with untreated control culture preparations, as measured by means of histologic autoradiography. Comparisons were made with clinical responses, ie, complete response, partial response, or no response. PATIENTS: The study was carried out in patients with head and neck cancers and comprised 21 patients with squamous-cell carcinoma, three patients with other carcinomas, and two patients with sarcoma. RESULTS: Ten of 12 patients with in vitro-sensitive tumors had either complete or partial response clinically. The overall accuracy of the SSHDRA was 74% in this correlative clinical trial; the predictive-positive value was 83%, the sensitivity was 71%, and the specificity was 78%. Seven of 11 patients with in vitro-resistant tumors demonstrated no response for a predictive-negative value of 64%. CONCLUSIONS: We conclude that the SSHDRA shows a high correlation for tumors that demonstrate both in vivo drug resistance and sensitivity. The in vitro-like maintenance of three-dimensional tissue architecture of the tumors in histoculture probably contributes to high clinical predictivity of drug response of the SSHDRA. The data support further comparisons to determine the clinical usefulness of the SSHDRA for identifying complete and partial responders to chemotherapy.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Drug Monitoring/methods , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Cisplatin/therapeutic use , Drug Screening Assays, Antitumor/methods , Humans , In Vitro Techniques , Predictive Value of Tests , Remission Induction , Sarcoma/drug therapy , Sarcoma/pathology , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL