Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Virchows Arch ; 470(5): 545-552, 2017 May.
Article in English | MEDLINE | ID: mdl-28213807

ABSTRACT

We examined somatostatin receptor type 2A (SSTR2A) expression in primary and metastatic small intestinal neuroendocrine tumors (SI-NETs). We retrieved 156 liver metastases from 26 patients (10 males, 16 females) who had two or more liver lesions resected. A representative formalin-fixed paraffin-embedded section of tumor tissue from each liver metastasis and from the primary tumor, when available, were immunohistochemically stained for SSTR2A. SSTR2A expression was evaluated by the Her2/neu-scoring system and the scoring system proposed by Volante et al. Based on the Her2/neu-scoring system, moderate to strong SSTR2A expression was observed in 121 of 156 (78%) liver metastases. In 15 (58%) subjects, all liver metastases showed moderate to strong SSTR2A expression, whereas in 11 (42%) one or more liver tumors had weak or no expression. Of the 16 stained primaries, 11 (69%) showed heterogeneous SSTR2A expression. The corresponding liver metastases showed only weak to no expression in one, moderate to strong in five, and both weak to no and moderate to strong expression in five of the 11 cases. Using the Volante scoring system, no tumor was scored 0 (0%), two were scored 1 (1%), 38 were scored 2 (24%), and 116 were scored 3 (74%). No statistically significant association was observed between SSTR2A expression and Ki67 index (p = 0.56). Fifteen of 18 (83%) metastatic tumors with a Ki67 index >20% showed moderate to strong SSTR2A. Most liver tumors with weak SSTR2A expression or an IHC score of 2 were detected by OctreoScan. SSTR2A expression in liver metastases of SI-NETs can be variable, even between lesions in the same patient. Expression in metastatic lesions is not always similar to that in the primary tumor. SSTR2A expression is not associated with the Ki67 index.


Subject(s)
Biomarkers, Tumor/analysis , Intestinal Neoplasms/secondary , Liver Neoplasms/secondary , Neuroendocrine Tumors/pathology , Receptors, Somatostatin/biosynthesis , Adult , Aged , Female , Humans , Immunohistochemistry , Male , Middle Aged , Receptors, Somatostatin/analysis , Young Adult
2.
Am J Clin Pathol ; 143(3): 398-404, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25696798

ABSTRACT

OBJECTIVES: We examined Ki-67 heterogeneity within single and between synchronous liver metastases of small intestine neuroendocrine tumors. METHODS: There were 27 patients (10 men and 17 women) with two or more liver metastases. The Ki-67 index was used to classify the tumors into World Health Organization grade 1, 2, or 3. The association between Ki-67 heterogeneity and tumor size of liver metastases was analyzed. Correlation of tumor grade with patient survival was also evaluated. RESULTS: Primary tumors from 20 patients were graded, including 17 grade 1 and three grade 2. A total of 188 liver metastases were resected, including 122 (65%) grade 1, 47 (25%) grade 2, and 19 (10%) grade 3. The highest tumor grade was grade 1 in 10 (37%), grade 2 in nine (33%), and grade 3 in eight (30%) patients. Patients with one or more grade 3 liver lesions had a shorter progression-free survival compared with those with grade 1/2 tumors (P < .001). A positive association was found between tumor size and Ki-67 index (P = .04), as well as between tumor size and intratumoral Ki-67 heterogeneity (P < .001). CONCLUSIONS: Intratumoral and intertumoral Ki-67 heterogeneity is common and positively correlated with tumor size. The presence of one or more grade 3 liver lesions predicts a worse prognosis.


Subject(s)
Biomarkers, Tumor/metabolism , Intestinal Neoplasms/pathology , Ki-67 Antigen/metabolism , Liver Neoplasms/secondary , Neuroendocrine Tumors/secondary , Adult , Aged , Demography , Disease Progression , Disease-Free Survival , Education, Medical, Continuing , Female , Humans , Intestinal Neoplasms/classification , Intestinal Neoplasms/mortality , Intestine, Small/pathology , Liver/pathology , Liver Neoplasms/classification , Liver Neoplasms/mortality , Male , Middle Aged , Neoplasm Grading , Neuroendocrine Tumors/classification , Neuroendocrine Tumors/mortality , Prognosis , World Health Organization , Young Adult
3.
J Natl Compr Canc Netw ; 11(6): 688-723, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23744868

ABSTRACT

Primary bone cancers are extremely rare neoplasms, accounting for fewer than 0.2% of all cancers. The evaluation and treatment of patients with bone cancers requires a multidisciplinary team of physicians, including musculoskeletal, medical, and radiation oncologists, and surgeons and radiologists with demonstrated expertise in the management of these tumors. Long-term surveillance and follow-up are necessary for the management of treatment late effects related to surgery, radiation therapy, and chemotherapy. These guidelines discuss the management of chordoma, giant cell tumor of the bone, and osteosarcoma.


Subject(s)
Bone Neoplasms/diagnosis , Bone Neoplasms/therapy , Humans , Neoplasm Staging
4.
Cancer Sci ; 103(12): 2173-80, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23035734

ABSTRACT

Despite reports of sex steroid receptor and COX2 expression in desmoid-type fibromatosis, responses to single agent therapy with anti-estrogens and non-steroidal anti-inflammatory drugs are unpredictable. Perhaps combination pharmacotherapy might be more effective in desmoid tumors that co-express these targets. Clearly, further understanding of the signaling pathways deregulated in desmoid tumors is essential for the development of targeted molecular therapy. Transforming growth factor-ß (TGFß) and bone morphogenetic proteins (BMP) are important regulators of fibroblast proliferation and matrix deposition, but little is known about the TGFß superfamily in fibromatosis. A tissue microarray representing 27 desmoid tumors was constructed; 14 samples of healing scar and six samples of normal fibrous tissue were included for comparison. Expression of selected receptors and activated downstream transcription factors of TGFß family signaling pathways, ß-catenin, sex steroid hormone receptors and COX2 were assessed using immunohistochemistry; patterns of co-expression were explored via correlational statistical analyses. In addition to ß-catenin, immunoreactivity for phosphorylated SMAD2/3 (indicative of active TGFß signaling) and COX2 was significantly increased in desmoid tumors compared with healing scar and quiescent fibrous tissue. Low levels of phosphorylated SMAD1/5/8 were detected in only a minority of cases. Transforming growth factor-ß receptor type 1 and androgen receptor were expressed in both desmoid tumors and scar, but not in fibrous tissue. Estrogen receptor-ß was present in all cases studied. Transforming growth factor-ß signaling appears to be activated in desmoid-type fibromatosis and phosphorylated SMAD2/3 and COX2 immunoreactivity might be of diagnostic utility in these tumors. Given the frequency of androgen receptor, estrogen receptor-ß and COX2 co-expression in desmoid tumors, further assessment of the efficacy of combination pharmacotherapy using hormonal agonists/antagonists together with COX2 inhibitors should be considered.


Subject(s)
Cyclooxygenase 2/metabolism , Estrogen Receptor beta/metabolism , Fibromatosis, Aggressive/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Adolescent , Adult , Bone Morphogenetic Proteins/metabolism , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Smad2 Protein/genetics , Smad2 Protein/metabolism , beta Catenin/metabolism
5.
Tumour Biol ; 33(5): 1639-44, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22623071

ABSTRACT

Expression of the p63 tumor suppressor protein has been reported in the mononuclear stromal cells of giant cell tumor of the bone, which may represent osteoblast-precursor cells. Only a limited number of osteoblastic tumors have been studied for p63 expression thus far. We therefore examined whether p63 may serve as a marker for osteoblastic differentiation in osteosarcomas or as a differential diagnostic marker to distinguish osteoblastoma from osteosarcoma. Immunohistochemical stains for p63 were performed on a tissue microarray containing 71 chemotherapy naïve biopsy samples of osteosarcoma, 21 whole sections of osteosarcoma, and 8 osteoblastomas. Nuclear p63 was detected in seven of eight osteoblastomas but was restricted to stromal cells within primitive, immature-appearing areas of osteoid deposition. Although only 7 of 71 (10 %) biopsy samples of osteosarcoma represented on the tissue microarray were positive for p63, 7 of 21 (33 %) osteosarcomas were positive when whole tissue sections were evaluated. Although p63 is detected in most osteoblastomas, it is also observed in a significant subset of osteosarcomas, severely limiting its utility in distinguishing between benign and malignant osteoblastic tumors. The relatively low prevalence of p63 expression in osteosarcoma would also seem to preclude its use as a marker of osteoblastic differentiation in skeletal sarcomas.


Subject(s)
Bone Neoplasms/metabolism , Osteoblastoma/metabolism , Osteosarcoma/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Adolescent , Adult , Biomarkers, Tumor , Bone Neoplasms/genetics , Child , Female , Humans , Male , Middle Aged , Osteoblastoma/genetics , Osteosarcoma/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Young Adult
6.
Hum Pathol ; 43(10): 1711-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22520949

ABSTRACT

Despite reports of receptor tyrosine kinase activation in desmoid-type fibromatosis, therapeutic benefits of kinase inhibitor therapy are unpredictable. Variability in signal transduction or cellular kinases heretofore unevaluated in desmoid tumors may be responsible for these inconsistent responses. In either case, a better understanding of growth regulatory signaling pathways is necessary to assess the theoretical potential of inhibitor therapy. Immunohistochemical analysis of tyrosine kinases and activated isoforms of downstream signal transduction proteins was performed on a tissue microarray containing 27 cases of desmoid-type fibromatosis and 14 samples of scar; 6 whole sections of normal fibrous tissue were studied for comparison. Platelet-derived growth factor receptor, ß type, and focal adhesion kinase 1 were expressed in all desmoid tumors and healing scars but only 80% and 50% of nonproliferative fibrous tissue samples, respectively. Hepatocyte growth factor receptor was detected in 89% of desmoids and all scars tested, but not in any of the fibrous tissue samples. Epidermal growth factor receptor was detected in only 12% of desmoids and not in scar or fibrous tissue. Mast/stem cell growth factor receptor, receptor tyrosine-protein kinase erbB-2, and phosphorylated insulin-like growth factor 1 receptor/insulin receptor were negative in all study cases. Variable levels of phosphorylated downstream signal transduction molecules RAC-α/ß/γ serine/threonine-protein kinase, mitogen-activated protein kinase, and signal transducer and activator of transcription-3 were observed in desmoids (58%, 62%, and 67%), scar tissues (100%, 86%, and 86%), and fibrous tissue (33%, 17%, and 17%). These results indicate that tyrosine kinase signaling is active in both fibromatosis and healing scar, but not in most nonproliferating fibrous tissues. Although platelet-derived growth factor receptor, ß type, is expressed ubiquitously in desmoids, the kinases driving cell proliferation in desmoids remain unresolved.


Subject(s)
Fibroma/enzymology , Fibroma/pathology , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Infant , Male , Middle Aged , Tissue Array Analysis , Young Adult
7.
Hum Pathol ; 43(9): 1418-24, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22404950

ABSTRACT

The morphologic changes seen in desmoid-type fibromatosis after radiotherapy have not been well studied, and the degree of cytologic atypia, cellularity, mitotic activity, and lesional necrosis found in desmoid-type fibromatosis treated with ionizing radiation has not been thoroughly assessed. Therefore, we evaluated a series of primary and locally recurrent desmoid-type fibromatoses resected at variable intervals after radiation therapy (XRT) and compared their histopathologic and immunophenotypic features with paired pathologic specimens that were obtained before radiotherapy. The morphologic characteristics of desmoid-type fibromatoses resected before and after radiotherapy were not significantly different in 7 of the 8 cases studied. One case resected 191 months after XRT showed morphologic evidence of fibrosarcomatous transformation, with zonal necrosis, hypercellularity, severe nuclear atypia, and increased mitotic activity. No significant differences in the immunophenotype of desmoid-type fibromatosis were observed after XRT. In rare cases of recurrent desmoid-type fibromatosis, the differential diagnosis may include radiation-induced fibrosarcoma. Our data suggest that histomorphologic alterations attributable to the effects of ionizing radiation in desmoid-type fibromatosis are minimal. Therefore, the presence of cytologic features of malignancy in this setting warrants careful examination and consideration of the rare occurrence of postradiation sarcoma. Lesser degrees of nuclear atypia seen in isolation do not necessarily indicate a poor prognosis in irradiated desmoid-type fibromatosis.


Subject(s)
Fibromatosis, Aggressive/pathology , Neoplasm Recurrence, Local/pathology , Soft Tissue Neoplasms/pathology , Adolescent , Adult , Biomarkers, Tumor/metabolism , Child , Female , Fibromatosis, Aggressive/metabolism , Fibromatosis, Aggressive/radiotherapy , Humans , Male , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/radiotherapy , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/radiotherapy , Treatment Outcome
9.
J Clin Oncol ; 27(25): 4188-96, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19652065

ABSTRACT

PURPOSE: To evaluate the safety and efficacy of trabectedin in a phase II, open-label, multicenter, randomized study in adult patients with unresectable/metastatic liposarcoma or leiomyosarcoma after failure of prior conventional chemotherapy including anthracyclines and ifosfamide. PATIENTS AND METHODS: Patients were randomly assigned to one of two trabectedin regimens (via central venous access): 1.5 mg/m(2) 24-hour intravenous infusion once every 3 weeks (q3 weeks 24-hour) versus 0.58 mg/m(2) 3-hour IV infusion every week for 3 weeks of a 4-week cycle (qwk 3-hour). Time to progression (TTP) was the primary efficacy end point, based on confirmed independent review of images. RESULTS: Two hundred seventy patients were randomly assigned; 136 (q3 weeks 24-hour) versus 134 (qwk 3-hour). Median TTP was 3.7 months versus 2.3 months (hazard ratio [HR], 0.734; 95% CI, 0.554 to 0.974; P = .0302), favoring the q3 weeks 24-hour arm. Median progression-free survival was 3.3 months versus 2.3 months (HR, 0.755; 95% CI, 0.574 to 0.992; P = .0418). Median overall survival (n = 235 events) was 13.9 months versus 11.8 months (HR, 0.843; 95% CI, 0.653 to 1.090; P = .1920). Although somewhat more neutropenia, elevations in AST/ALT, emesis, and fatigue occurred in the q3 weeks 24-hour, this regimen was reasonably well tolerated. Febrile neutropenia was rare (0.8%). No cumulative toxicities were noted. CONCLUSION: Prior studies showed clinical benefit with trabectedin in patients with sarcomas after failure of standard chemotherapy. This trial documents superior disease control with the q3 weeks 24-hour trabectedin regimen in liposarcomas and leiomyosarcomas, although the qwk 3-hour regimen also demonstrated activity relative to historical comparisons. Trabectedin may now be considered an important new option to control advanced sarcomas in patients after failure of available standard-of-care therapies.


Subject(s)
Anthracyclines/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/therapeutic use , Dioxoles/administration & dosage , Drug Resistance, Neoplasm , Ifosfamide/therapeutic use , Leiomyosarcoma/drug therapy , Liposarcoma/drug therapy , Tetrahydroisoquinolines/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/adverse effects , Australia , Dioxoles/adverse effects , Disease-Free Survival , Drug Administration Schedule , Europe , Female , Humans , Infusions, Intravenous , Kaplan-Meier Estimate , Leiomyosarcoma/secondary , Liposarcoma/secondary , Male , Middle Aged , Neoplasm Staging , North America , Proportional Hazards Models , Risk Assessment , Tetrahydroisoquinolines/adverse effects , Time Factors , Trabectedin , Treatment Failure , Young Adult
10.
J Clin Oncol ; 27(10): 1706-11, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19224843

ABSTRACT

The American Society of Clinical Oncology (ASCO) developed its own test -- the Medical Oncology In-Training Examination (MedOnc ITE) -- as a tool to assess trainees' knowledge of the clinical oncology subspecialty, establish consistency in educational standards across training programs, identify areas of strength and weakness in individual programs, and stimulate intraprogrammatic reading and discussion. The Accreditation Council for Graduate Medical Education Outcome Project provided additional incentive for ASCO to develop an ITE. The examination was developed in 4 years. The concept of the examination and the budget were approved by the ASCO governing board. The National Board of Medical Examiners was selected to work with ASCO. Fellowship programs were contacted to determine if they had the information technology support to hold the examination. A blueprint for the examination was developed. The test format, including the number of questions and the selection of case-based single best answers, was determined. Physician volunteers to write the questions were solicited from among program directors, various ASCO committees, and disease experts. A workshop was held to teach volunteers how to write proper case-based questions. From this pool, a smaller group of physicians was selected to develop the test and review all test questions. The final examination was developed and administered in February 2008, with scores provided to fellows and program directors in April 2008. Feedback received after the examination will be helpful for developing future MedOnc ITEs. The process ASCO went through to develop the MedOnc ITE serves as a model for other subspecialties interested in developing their own ITEs.


Subject(s)
Education, Medical, Graduate/methods , Educational Measurement/methods , Medical Oncology , Education, Medical, Graduate/standards , Fellowships and Scholarships , Humans , Societies, Medical
11.
Clin Cancer Res ; 12(9): 2834-40, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16675578

ABSTRACT

PURPOSE: Microtubules play a critical role in many cellular functions, including cell division and mitosis. ABT-751 is a novel sulfonamide antimitotic that binds to the colchicine site on beta-tubulin that leads to a block in the cell cycle at the G2M phase, resulting in cellular apoptosis. ABT-751 was investigated in this phase 1 trial designed to assess its maximum tolerated dose (MTD), dose-limiting toxicity (DLT), tolerability, and pharmacokinetics. EXPERIMENTAL DESIGN: ABT-751 was administered on a daily (q.d.) or twice daily (b.i.d.) oral schedule for 7 days every 3 weeks to 39 patients with refractory solid tumors. Toxicity was monitored weekly. Plasma and urine ABT-751 and metabolite pharmacokinetics were determined. RESULTS: The MTD for the q.d. schedule was 250 mg/d. DLTs during cycle 1 were abdominal pain, constipation, and fatigue. The MTD on the b.i.d. schedule was 150 mg. Cycle 1 of therapy with the 175 mg b.i.d. schedule was tolerated without DLT. However, six of seven patients reported grade 3 toxicity (ileus, constipation, abdominal pain, or fatigue), which occurred in cycle 2 or 3. ABT-751 was absorbed after oral administration with an overall mean T(max) of about 2 hours. The pharmacokinetics of ABT-751 were dose-proportional and time-independent. There was minimal accumulation of ABT-751 after multiple q.d. and b.i.d. doses. Efficacious concentrations, as determined from preclinical models (0.5-1.5 microg/mL), were achieved in all subjects. ABT-751 metabolism occurred primarily by glucuronidation and sulfation. No complete or partial tumor responses were noted, but one patient had a minor response, and four patients had stable disease lasting at least 6 months. CONCLUSIONS: The MTD and recommended phase 2 doses for ABT-751 were 250 mg q.d. and 150 mg b.i.d. on a 7-day schedule given every 3 weeks, due to subsequent cycle toxicities at 175 mg b.i.d. dosing. Toxicities were abdominal pain, constipation, and neuropathy.


Subject(s)
Neoplasms/blood , Sulfonamides/pharmacokinetics , Sulfonamides/toxicity , Administration, Oral , Adult , Aged , Biological Availability , Cell Division/drug effects , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Neoplasms/urine , Safety , Sulfonamides/therapeutic use
12.
Am J Clin Oncol ; 29(2): 109-15, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16601426

ABSTRACT

OBJECTIVES: Determine the toxicity, tolerability, and pharmacokinetics of SU5416, a vascular endothelial growth factor receptor-2 (VEGFR-2) tyrosine kinase inhibitor, coadministered with bolus 5-fluorouracil (5-FU), leucovorin, and irinotecan (IFL) in untreated patients with metastatic colorectal cancer. METHODS: SU5416 (85 or 145 mg/m2) was administered twice weekly throughout a 6-week period along with standard IFL (4 weeks on/2 weeks off). Plasma samples were assayed for SU5416, irinotecan, and SN-38 by reverse-phase HPLC. Contrast enhanced, color Doppler sonography was performed on patients at the MTD to identify changes in tumor perfusion. RESULTS: Eleven patients received treatment with SU5416 85 mg/m2 (n = 5) or 145 mg/m2 (n = 6). At 85 mg/m2, no DLTs were observed. At 145 mg/m2, grade 3 diarrhea and vomiting were observed during cycle 1; other grade 3 toxicities included fatigue, nausea, anorexia, anemia, pain, urinary retention, and hypertension. The pharmacokinetics of irinotecan and SN-38 were not altered by coadministration of SU5416. SU5416 pharmacokinetics were not altered by IFL. Contrast-enhanced, color Doppler sonography was performed on 2 patients and demonstrated reduced tumor perfusion after treatment in a patient who responded to treatment and increased perfusion in a patient who developed progressive disease. Three patients (27%) had confirmed partial responses, 2 patients (18%) had unconfirmed partial responses, and 4 patients (36%) had stable disease. CONCLUSIONS: Twice weekly SU5416 can be administered with bolus IFL without unexpected toxicities or altering the pharmacokinetic behavior of the administered drugs. Changes in tumor blood perfusion can be detected by contrast-enhanced, color Doppler sonography. The further development of SU5416 was halted before this study was completed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Colorectal Neoplasms/pathology , Drug Administration Schedule , Female , Fluorouracil/administration & dosage , Humans , Indoles/administration & dosage , Infusions, Intravenous , Injections, Intravenous , Irinotecan , Leucovorin/administration & dosage , Male , Middle Aged , Neoplasm Metastasis , Pyrroles/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/physiology
14.
Cancer Chemother Pharmacol ; 55(6): 609-16, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15723220

ABSTRACT

BACKGROUND: Aprepitant is a selective neurokinin-1 receptor antagonist that is effective for the prevention of nausea and vomiting caused by highly emetogenic chemotherapy. In vitro, aprepitant is a moderate inhibitor of the CYP3A4 enzyme, which is involved in the clearance of several chemotherapeutic agents. In this study we examined the potential for aprepitant to affect the pharmacokinetics and toxicity of intravenously administered docetaxel, a chemotherapeutic agent that is primarily metabolized by CYP3A4. METHODS: A total of 11 cancer patients (4 male, 7 female, aged 50-68 years) were enrolled in this multicenter, randomized, open-label, two-period, crossover study. Patients received a single infusion of docetaxel monotherapy, 60-100 mg/m(2), on two occasions at least 3 weeks apart. During one of the cycles (treatment A), patients received docetaxel alone. During the alternate cycle (treatment B), they also received aprepitant 125 mg orally 1 h prior to docetaxel infusion (day 1), and a single oral dose of aprepitant 80 mg on days 2 and 3. The pharmacokinetic profile of docetaxel was assessed over 30 h following docetaxel infusion. Blood counts were monitored on days 1, 4, 7, and 14. RESULTS: Ten patients completed the study. Concomitant administration of aprepitant did not cause any statistically or clinically significant changes in docetaxel pharmacokinetics. Values for docetaxel alone (treatment A) versus docetaxel with aprepitant (treatment B) were as follows: geometric mean AUC(0-last) was 3.26 vs 3.17 microg h/ml (P>0.25; ratio B/A 0.97); geometric mean AUC(0-infinity) 3.51 vs 3.39 microg h/ml (P>0.25; ratio B/A 0.96); geometric mean C(max) was 3.53 vs 3.37 microg/ml (P>0.25; ratio B/A 0.95); and geometric mean plasma clearance was 23.3 vs 24.2 l/h/m(2) (P>0.25; ratio B/A 1.04). The corresponding harmonic mean half-life values were 10.1 and 8.5 h. The two treatment regimens had similar tolerability profiles; the median absolute neutrophil count nadirs were 681/mm(3) during treatment with docetaxel alone and 975/mm(3) during aprepitant coadministration. CONCLUSIONS: Aprepitant had no clinically significant effect on either the pharmacokinetics or toxicity of standard doses of docetaxel in cancer patients. Aprepitant at clinically recommended doses may have a low potential to affect the pharmacokinetics of intravenous chemotherapeutic agents metabolized by CYP3A4.


Subject(s)
Antiemetics/adverse effects , Antineoplastic Agents, Phytogenic/pharmacokinetics , Morpholines/adverse effects , Neoplasms/drug therapy , Taxoids/pharmacokinetics , Administration, Oral , Aged , Antiemetics/administration & dosage , Antiemetics/therapeutic use , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/therapeutic use , Aprepitant , Cross-Over Studies , Docetaxel , Female , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate/drug effects , Middle Aged , Morpholines/administration & dosage , Morpholines/therapeutic use , Nausea/prevention & control , Neoplasms/metabolism , Taxoids/adverse effects , Taxoids/therapeutic use
15.
Clin Cancer Res ; 10(3): 909-15, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871966

ABSTRACT

PURPOSE: Prinomastat is a matrix metalloprotease (MMP) inhibitor with selectivity for MMPs 2, 3, 9, 13, and 14. Inhibition of these MMPs has been postulated to block tumor invasion and metastasis. This Phase I, dose-escalation study was designed to evaluate the acute and chronic toxicities of various doses of prinomastat and to determine prinomastat pharmacokinetics. EXPERIMENTAL DESIGN: Seventy-five patients with advanced cancer were given 1, 2, 5, 10, 25, 50, or 100 mg prinomastat orally twice daily until tumor progression or development of significant toxicities. Prinomastat pharmacokinetics were measured on day 29 of therapy. RESULTS: The primary toxicities identified were joint and muscle-related pain, which were generally reversible with treatment rest and/or dose reduction. No dose-limiting toxicities were noted within the first 4 weeks of treatment, but grade 2-3 arthralgias and myalgias were noted 2-3 months after initiation of therapy in >25% of patients at doses >25 mg twice a day. The frequency and severity of symptoms were dose related. Plasma prinomastat concentrations greater than the K(i) for MMPs 2 and 9 were achieved at all of the dose levels. CONCLUSIONS: Doses of 5-10 mg bid were recommended for additional trials, because this dose range was well tolerated for a treatment duration of at least 3 months and achieves trough plasma concentrations 10-100-fold greater than the K(i) (in vitro inhibition constant) for the targeted MMPs (2 and 9).


Subject(s)
Enzyme Inhibitors/therapeutic use , Matrix Metalloproteinase Inhibitors , Organic Chemicals/therapeutic use , Adult , Aged , Aged, 80 and over , Alkaline Phosphatase/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Area Under Curve , Disease Progression , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Kinetics , Male , Maximum Tolerated Dose , Middle Aged , Organic Chemicals/pharmacokinetics , Organic Chemicals/pharmacology , Time Factors
16.
Clin Cancer Res ; 10(2): 468-75, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14760067

ABSTRACT

PURPOSE: This study was performed to determine the maximum tolerated dose, dose-limiting toxicities, and pharmacokinetics of brostallicin, a nonalkylating DNA minor groove binder and a synthetic derivative of distamycin A, given as a weekly i.v. infusion. EXPERIMENTAL DESIGN: Using an accelerated dose escalation design, patients with advanced solid tumor malignancies were treated with brostallicin administered as a 10-min i.v. infusion on days 1, 8, and 15 of a 28-day cycle. The starting dose of brostallicin was 0.3 mg/m(2)/week. To study the pharmacokinetic behavior of brostallicin, serial blood samples were obtained before and after the first and last infusions during cycle 1, and in cycles 2 and 4 in a limited number of patients. RESULTS: Fourteen patients received 32 complete cycles of brostallicin. Dose-limiting toxicity was febrile neutropenia and was observed in 3 of 5 patients treated at 4.8 mg/m(2)/week. The maximum tolerated dose and recommended Phase II dose was 2.4 mg/m(2)/week. The mean +/- SD terminal half-life at the maximum tolerated dose was 4.6 +/- 4.1 h. There was moderate distribution of brostallicin into tissues, and the clearance was approximately 20% of the hepatic blood flow. The area under the concentration time curve(0- infinity ) of brostallicin increased in a dose-linear fashion. No significant relationship was observed between any plasma pharmacokinetic parameter and clinical toxicities. There were no objective responses during the trial, but 5 patients had stable disease after two cycles of treatment. CONCLUSIONS: The dose-limiting toxicity of weekly brostallicin was neutropenia. Systemic exposure increases linearly with dose. The recommended dose for Phase II studies is 2.4 mg/m(2) on days 1, 8, and 15 of a 28-day cycle.


Subject(s)
Guanidines/pharmacokinetics , Guanidines/therapeutic use , Neoplasms/drug therapy , Pyrroles/pharmacokinetics , Pyrroles/therapeutic use , Aged , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Area Under Curve , Calibration , DNA Damage , Female , Humans , Kinetics , Male , Middle Aged , Models, Chemical , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...