Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Cancer Chemother Pharmacol ; 67(4): 809-12, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20563581

ABSTRACT

PURPOSE: MP470 is a multi-targeted tyrosine kinase inhibitor with potent activity against mutant c-Kit, PDGFRα, Flt3, c-Met and c-Ret that is being evaluated as an anticancer agent. The plasma and cerebrospinal fluid (CSF) pharmacokinetics of MP470 were studied in a non-human primate model that is highly predictive of CSF penetration in humans. METHODS: Oral MP470, 300 mg, was administered to four non-human primates. Serial samples of blood were collected from four animals and CSF samples from three animals for pharmacokinetic studies. Plasma and CSF concentrations were measured using an LC-MS/MS assay. Both model-independent and model-dependent methods were used to analyze the pharmacokinetic data. RESULTS: Following a one-time oral dose of 300 mg, the MP470 plasma area under the curve (AUC) was 1,690 ± 821 nM h (mean ± SD). The half-life of MP470 in the plasma was 11.0 ± 3.4 h. There was no measurable MP470 in the CSF. CONCLUSIONS: Although CSF penetration is minimal, MP470 has demonstrated potent activity against cancer cell lines in vitro and in vivo, and further clinical investigation is warranted.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents/cerebrospinal fluid , Area Under Curve , Chromatography, Liquid , Half-Life , Macaca mulatta , Male , Models, Biological , Piperazines , Protein Kinase Inhibitors/cerebrospinal fluid , Pyrimidines/cerebrospinal fluid , Tandem Mass Spectrometry , Thiourea
2.
Neuropathol Appl Neurobiol ; 33(1): 67-76, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17239009

ABSTRACT

Survivin, a member of the inhibitor of apoptosis protein family, is implicated in the dysregulation of apoptosis in human cancers. Survivin and survivin-deltaEx3, one of its two alternatively spliced isoforms, confer anti-apoptotic activities in human tumours, while survivin-2B antagonizes such anti-apoptotic properties. The current study was undertaken to examine the mRNA expression of survivin isoforms and their correlation with clinical staging and outcome in 20 medulloblastoma (MB) tumours, three MB cell lines and normal brain tissues (a foetal and an adult cerebellum) by densitometry scanning of 32p-dCTP incorporated reverse transcription polymerase chain reaction (RT-PCR) products and quantitative real-time PCR. Our results showed that the normal adult brain only expressed low levels of survivin-deltaEx3 mRNA, while the foetal brain expressed all three isoforms, with wild-type survivin as the dominant transcript. All three survivin isoforms were detected in all the MB cell lines and tumours analysed. Immunohistochemical staining also demonstrated survivin protein expressions in all five paraffin-embedded MBs, with predominant nuclear localization. Although overexpressions of survivin were not associated with the presence of metastatic MB or tumour histological subtypes, elevated expressions of survivin-deltaEx3 were significantly associated with progressive/recurrent tumours (P-value = 0.024). Our data demonstrated that overexpression of survivin mRNA is a common feature in MBs, may contribute to their anti-apoptosis properties and clinical behaviours, and predicts a poor clinical outcome, independent of clinical staging or tumour histology.


Subject(s)
Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Microtubule-Associated Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Adolescent , Cell Line, Tumor , Cerebellar Neoplasms/genetics , Child , Child, Preschool , Female , Gene Expression Regulation, Neoplastic , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Immunohistochemistry , Infant , Inhibitor of Apoptosis Proteins , Isomerism , Male , Medulloblastoma/genetics , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Neoplasm Staging , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Survivin , Treatment Outcome
3.
Pediatr Blood Cancer ; 46(1): 50-5, 2006 Jan.
Article in English | MEDLINE | ID: mdl-15768380

ABSTRACT

BACKGROUND: The objectives of this study were to determine the maximum tolerated dose (MTD), dose-limiting toxicities (DLTs), pharmacokinetics, and anti-tumor effect of irinotecan in pediatric patients with recurrent or refractory malignancies. PROCEDURE: Twenty-three patients between 1 and 21 years of age, with a solid tumor refractory to standard therapy or for which there was no standard therapy were enrolled. Irinotecan was administered over 90 min weekly 4x, every 6 weeks. The initial dose level was 125 mg/m(2)/day, with subsequent escalations to 160 and 200 mg/m(2)/day. A MTD was defined in heavily-pretreated and less-heavily-pretreated (< or =2 prior chemotherapy regimens, no prior bone marrow transplantation, and no central axis radiation) patients. Pharmacokinetic studies were also performed. RESULTS: Neutropenia and diarrhea were the DLTs in heavily pretreated patients; the MTD was 125 mg/m(2)/day. Neutropenia was the DLT in less-heavily pretreated; the MTD was 160 mg/m(2)/day. Five patients had stable disease for two to four cycles including one patient each with rhabdomyosarcoma, Ewing sarcoma, neuroblastoma, and two patients with ependymoma. Irinotecan clearance was greater that that previously reported for children receiving high dose irinotecan. CONCLUSIONS: The recommended phase II dose of irinotecan administered weekly 4x, every 6 weeks in children with solid tumors is 125 mg/m(2)/dose for heavily pretreated patients and 160 mg/m(2)/dose for less heavily pretreated patients.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Neoplasms/drug therapy , Adolescent , Adult , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Child , Child, Preschool , Drug Administration Schedule , Female , Humans , Irinotecan , Male , Maximum Tolerated Dose
4.
J Clin Oncol ; 22(19): 3916-21, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15459213

ABSTRACT

PURPOSE: We performed a phase I trial of intrathecal (IT) liposomal cytarabine (DepoCyt; Enzon Pharmaceuticals, Piscataway, NJ and SkyePharma Inc, San Diego, CA) to determine the maximum-tolerated dose, the dose-limiting toxicities, and the plasma and CSF pharmacokinetics of IT lipsomal cytarabine in children >/= 3 years of age with advanced meningeal malignancies. PATIENTS AND METHODS: Eighteen assessable patients received IT liposomal cytarabine through either an indwelling ventricular access device or via lumbar puncture. Liposomal cytarabine was given once every 2 weeks during induction, once every 4 weeks during consolidation, and once every 8 weeks during the maintenance phase of treatment. The initial dose was 25 mg, with subsequent escalations to 35 and 50 mg. CSF pharmacokinetic samples were obtained in a subset of patients. RESULTS: Arachnoiditis, characterized by fever, headache, nausea, vomiting, and back pain was noted in the first two patients at the 25 mg dose level. Therefore, subsequent patients were treated with dexamethasone, beginning the day of liposomal cytarabine administration and continuing for 5 days. Headache (grade 3) was dose limiting in two of eight patients enrolled at the 50 mg dose level. Eight of the 14 patients assessable for response demonstrated evidence of benefit manifest as prolonged disease stabilization or response. CONCLUSION: The maximum-tolerated dose and recommended phase II dose of liposomal cytarabine in patients between the ages of 3 and 21 years is 35 mg, administered with dexamethasone (0.15 mg/kg/dose, twice a day for 5 days). A phase II trial of IT liposomal cytarabine in children with CNS leukemia in second or higher relapse is in development.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Cytarabine/administration & dosage , Meningeal Neoplasms/drug therapy , Adolescent , Adult , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/pharmacokinetics , Catheters, Indwelling , Child , Child, Preschool , Cytarabine/adverse effects , Cytarabine/pharmacokinetics , Drug Delivery Systems , Female , Glioma/drug therapy , Glioma/metabolism , Humans , Injections, Spinal , Leukemia/drug therapy , Liposomes , Male , Maximum Tolerated Dose , Medulloblastoma/drug therapy , Medulloblastoma/metabolism , Meningeal Neoplasms/metabolism , Spinal Puncture
5.
Cancer Chemother Pharmacol ; 53(6): 482-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14999430

ABSTRACT

PURPOSE: To determine the maximum tolerated dose and the toxicity profile of the PDGF receptor pathway inhibitor SU101 in pediatric patients with refractory solid tumors, and to define the plasma pharmacokinetics of SU101 and its active metabolite SU0020 in children. EXPERIMENTAL DESIGN: Patients between 3 and 21 years of age with CNS malignancy, neuroblastoma, or sarcoma refractory to standard therapy were eligible. The starting dose of SU101 was 230 mg/m(2) per day administered as a 96-h continuous infusion every 21 days. Blood for pharmacokinetic analysis was obtained during the first cycle. RESULTS: Entered into the trial were 27 patients, and 24 were fully evaluable for toxicity. Dose-limiting central nervous system toxicity was observed in two patients at the 440 mg/m(2) per day dose level. Non-dose-limiting toxicities included nausea, vomiting, headache, fatigue, abdominal discomfort, diarrhea, pruritus, anorexia, constipation, and paresthesias. There were no complete or partial responses. One patient with rapidly progressive desmoplastic small round-cell tumor experienced symptomatic improvement and prolonged stable disease. Steady-state concentrations of SU101 were rapidly achieved and proportional to dose. The concentration of SU0020 was 100- to 1000-fold greater than that of SU101. The median clearance of SU0020 was 0.19 l/day per m(2) and its terminal elimination half-life was 14 days. CONCLUSIONS: SU101 administered on this schedule was generally well tolerated. The maximum tolerated dose of SU101 is 390 mg/m(2) per day for 4 days repeated every 3 weeks. The neurotoxicity observed at the 440 mg/m(2) per day dose level suggests that patients receiving repetitive cycles must be monitored closely, as SU0020 may accumulate over time.


Subject(s)
Antineoplastic Agents/therapeutic use , Central Nervous System Neoplasms/drug therapy , Isoxazoles/therapeutic use , Adolescent , Adult , Aniline Compounds/blood , Antineoplastic Agents/pharmacokinetics , Central Nervous System Neoplasms/metabolism , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Isoxazoles/pharmacokinetics , Leflunomide , Magnetic Resonance Imaging , Male , Nitriles/blood , Tomography, X-Ray Computed , Treatment Outcome
6.
Oncologist ; 6(6): 506-16, 2001.
Article in English | MEDLINE | ID: mdl-11743213

ABSTRACT

Camptothecin analogs, agents that target the intranuclear enzyme topoisomerase I, represent a promising new class of anticancer drugs for the treatment of childhood cancer. In preclinical studies, camptothecins, such as topotecan and irinotecan, are highly active against a variety of pediatric malignancies including neuroblastomas, rhabdomyosarcomas, gliomas, and medulloblastomas. In this paper, we review the status of completed and ongoing clinical trials and pharmacokinetic studies of camptothecin analogs in children. These and future planned studies of this novel class of cytotoxic agents are critical to defining the ultimate role of topoisomerase I poisons in the treatment of childhood cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Topoisomerase I Inhibitors , Topotecan/therapeutic use , Adolescent , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/adverse effects , Camptothecin/pharmacokinetics , Child , Child, Preschool , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Humans , Infant , Irinotecan , Topotecan/adverse effects , Topotecan/pharmacokinetics
7.
Curr Opin Oncol ; 13(3): 154-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11307057

ABSTRACT

Medulloblastoma, the most common primary malignant brain tumor in children, is a radiosensitive and chemosensitive tumor. Nevertheless, medulloblastoma remains a management challenge for the clinical oncologist, because the optimal sequence and dosage for each treatment modality has not yet been defined. In addition, effective management strategies for medulloblastoma may result in profound neuroendocrine and neuropsychologic sequelae. In this article, we review the clinical and biologic prognostic factors for classifying medulloblastoma, current strategies for the management of this disease, and potential strategies to prevent or minimize long-term treatment sequelae.


Subject(s)
Cerebellar Neoplasms/therapy , Medulloblastoma/therapy , Cerebellar Neoplasms/diagnosis , Child , Combined Modality Therapy , Humans , Medulloblastoma/diagnosis , Prognosis , Treatment Outcome
8.
Crit Rev Oncol Hematol ; 37(3): 227-36, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11248578

ABSTRACT

An unforeseen consequence of improved disease-free survival in many hematologic and solid tumor malignancies has been an increase in the incidence of disease recurrence in the leptomeninges. The recognition of the central nervous system (CNS) as a unique 'sanctuary' site has resulted in the development of therapeutic strategies specifically directed at the leptomeninges. Although therapeutic strategies have been successful in the prevention and treatment of CNS leukemia, there are still a paucity of therapeutic options for patients with neoplastic meningitis due to solid tumors or recurrent CNS leukemia. This article provides an overview of the pharmacology and toxicity of intrathecal agents that are commonly employed in the treatment and prevention of leptomeningeal disease, and describes new agents that are in the early stages of clinical development.


Subject(s)
Antineoplastic Agents/administration & dosage , Meninges/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Humans , Injections, Spinal , Leukemic Infiltration/prevention & control , Meningeal Neoplasms/prevention & control
9.
J Clin Oncol ; 19(3): 697-704, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11157020

ABSTRACT

PURPOSE: To improve the control of hyperuricemia in patients with leukemia or lymphoma, we tested a newly developed uricolytic agent, recombinant urate oxidase (SR29142; Rasburicase; Sanofi-Synthelabo, Inc, Paris, France), which catalyzes the oxidation of uric acid to allantoin, a highly water-soluble metabolite readily excreted by the kidneys. PATIENTS AND METHODS: We administered Rasburicase intravenously, at 0.15 or 0.20 mg/kg, for 5 to 7 consecutive days to 131 children, adolescents, and young adults with newly diagnosed leukemia or lymphoma, who either presented with abnormally high plasma uric acid concentrations or had large tumor cell burdens. Blood levels of uric acid, creatinine, phosphorus, and potassium were measured daily. The pharmacokinetics of Rasburicase, the urinary excretion rate of allantoin, and antibodies to Rasburicase were also studied. RESULTS: At either dosage, the recombinant enzyme produced a rapid and sharp decrease in plasma uric acid concentrations in all patients. The median level decreased by 4 hours after treatment, from 9.7 to 1 mg/dL (P =.0001), in the 65 patients who presented with hyperuricemia, and from 4.3 to 0.5 mg/dL (P =.0001) in the remaining 66 patients. Despite cytoreductive chemotherapy, plasma uric acid concentrations remained low throughout the treatment (daily median level, 0.5 mg/dL). The urinary excretion rate of allantoin increased during Rasburicase treatment, peaking on day 3. Serum phosphorus concentrations did not change significantly during the first 3 days of treatment, decreased significantly by day 4 in patients presenting with hyperuricemia (P =.0003), and fell within the normal range in all patients by 48 hours after treatment. Serum creatinine levels decreased significantly after 1 day of treatment in patients with or without hyperuricemia at diagnosis (P =.0003 and P =.02, respectively) and returned to normal range in all patients by day 6 of treatment. Toxicity was negligible, and none of the patients required dialysis. The mean plasma half-lives of the agent were 16.0 +/- 6.3 (SD) hours and 21.1 +/- 12.0 hours, respectively, in patients treated at dosages of 0.15 or 0.20 mg/kg. Seventeen of the 121 assessable patients developed antibodies to the enzyme. CONCLUSION: Rasburicase is safe and highly effective for the prophylaxis or treatment of hyperuricemia in patients with leukemia or lymphoma.


Subject(s)
Burkitt Lymphoma/complications , Lymphoma, B-Cell/complications , Lymphoma, Non-Hodgkin/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Urate Oxidase/therapeutic use , Uric Acid/blood , Adolescent , Burkitt Lymphoma/blood , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Humans , Infant , Lymphoma, B-Cell/blood , Lymphoma, Non-Hodgkin/blood , Male , Phosphorus/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Recombinant Proteins/blood , Recombinant Proteins/therapeutic use , Urate Oxidase/blood
10.
Comp Med ; 50(5): 486-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11099129

ABSTRACT

A nonhuman primate model comprising adult male rhesus monkeys (Macaca mulatta) with chronically indwelling subcutaneous central venous access devices provides a unique opportunity to determine plasma pharmacokinetics of new drugs such as anticancer and anti- retroviral agents. The central venous access we use is a low-profile, single-septum, titanium port that is attached to a radiopaque, indwelling catheter; the catheter is implanted in an internal jugular vein. A common complication following placement of the venous access device was migration of the catheter tip. We therefore modified the standard procedure by cutting the silicone catheter and introducing the rigid connector to secure the catheter to the vessel at the insertion site (approximately 9 to 13 cm from the distal end of the catheter). Prior to the use of the connector, three of five catheters migrated within 4 weeks after placement. In contrast, all 13 internal jugular catheters with connectors have remained patent without migration of the catheter tip. Therefore, incorporation of the catheter connector appears to have eliminated the problem of catheter migration.


Subject(s)
Catheters, Indwelling/veterinary , Jugular Veins/surgery , Macaca mulatta/surgery , Prosthesis Implantation/veterinary , Analgesics, Opioid/administration & dosage , Anesthetics, Inhalation/administration & dosage , Animals , Buprenorphine/administration & dosage , Cefazolin/administration & dosage , Cephalosporins/administration & dosage , Isoflurane/administration & dosage , Male , Penicillins/administration & dosage , Pharmaceutic Aids/administration & dosage , Povidone/administration & dosage , Prosthesis Implantation/methods
11.
J Pediatr Hematol Oncol ; 22(6): 506-9, 2000.
Article in English | MEDLINE | ID: mdl-11132217

ABSTRACT

PURPOSE: Pyrazoloacridine (PZA), a rationally synthesized deoxyribonucleic acid (DNA) binding agent that preferentially inhibits ribonucleic acid rather than DNA synthesis, is active against hypoxic and noncycling tumor cells and has greater in vitro activity against a broad range of human solid tumor lines than against the L1210 murine leukemia line. The Pediatric Oncology Group conducted a phase II study to determine the activity of PZA administered as a 3-hour infusion. PATIENTS AND METHODS: The activity of PZA was evaluated in patients with a variety of childhood solid tumors including rhabdomyosarcoma, Ewing sarcoma/peripheral neuroectodermal tumor, neuroblastoma, osteogenic sarcoma, Wilms tumor, or other solid tumors (excluding brain tumors). In addition to a standard three-stage design to test the drug's activity in each tumor type, a global stopping rule was used such that if no complete or partial responses (CR or PR) occurred in the first 35 patients (pooled across all strata except "other"), the study would be closed. RESULTS: A total of 47 patients were entered into the study. Myelosuppression was the primary toxicity. Severe nonhematologic toxicity was uncommon. Only one patient exhibited grade 3 neurologic toxicity (anxiety). No CRs or PRs were observed. CONCLUSION: Use of the global stopping criterion permitted early identification of lack of activity of PZA against childhood solid tumors.


Subject(s)
Acridines/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Pyrazoles/therapeutic use , Acridines/adverse effects , Adolescent , Adult , Antineoplastic Agents/adverse effects , Anxiety , Child , Child, Preschool , Humans , Infant , Patient Selection , Pyrazoles/adverse effects , Thrombocytopenia/chemically induced
12.
Ann Pharmacother ; 34(10): 1173-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11054987

ABSTRACT

BACKGROUND: The therapeutic effectiveness of chemotherapy is often limited by the inability to sustain cytotoxic concentrations at the tumor site. Cytarabine liposome injection (DepoCyt), a sterile, injectable suspension of the antimetabolite cytarabine, encapsulated into multivesicular, lipid-based particles, has been developed to improve the treatment of neoplastic meningitis (NM) through sustained release of cytarabine. OBJECTIVE: To review the pharmacokinetics, efficacy, and safety of intrathecal DepoCyt for the treatment of NM secondary to lymphoma or solid tumors. RESULTS: In preclinical and clinical studies, DepoCyt markedly extended the duration of tumor exposure to cytotoxic concentrations of cytarabine compared with administration of unbound cytarabine. Data from recent clinical studies demonstrate that DepoCyt improves complete response rates among patients with NM secondary to lymphoma. Trends in time to neurologic progression and median survival also favored DepoCyt over unbound cytarabine in these studies. Data have also been presented that suggest that patients with NM secondary to solid tumors benefit more from DepoCyt than from conventional treatment approaches. Chemical arachnoiditis (i.e., headache, fever, nausea, vomiting) was common in patients receiving DepoCyt, however, symptoms were manageable with oral dexamethasone. CONCLUSIONS: Encapsulation of cytarabine into liposomes for sustained release prolongs tumor exposure to cytotoxic concentrations of cytarabine, which may improve therapeutic efficacy in patients with NM secondary to lymphoma or solid tumors.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cytarabine/pharmacology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Cytarabine/administration & dosage , Cytarabine/therapeutic use , Delayed-Action Preparations , Humans
13.
Med Oncol ; 17(3): 151-62, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10962524

ABSTRACT

Neoplastic meningitis is an increasingly recognized complication of advanced metastatic cancer and, if left undiagnosed or untreated, is characterized by rapid neurologic deterioration and death. Thus, the diagnosis and treatment of neoplastic meningitis present challenges for the clinical oncologist. The diagnosis of neoplastic meningitis is based on clinical signs and symptoms, laboratory analysis of cerebrospinal fluid to determine cell count and cytology, and analysis of neuroimaging studies for evidence of leptomeningeal or cranial nerve enhancement. Once diagnosed, conventional treatment regimens may include radiotherapy combined with systemic or intrathecal chemotherapy, often with the antimetabolites cytarabine and/or methotrexate. However, the prognosis for neoplastic meningitis secondary to an underlying solid tumor or recurrent leukemia is poor with conventional treatment regimens. Therefore, novel agents for intrathecal administration, including DepoCyttrade mark, mafosfamide, and topotecan, or novel therapeutic approaches, including conjugated monoclonal antibodies and immunotoxins or gene therapy, are currently under investigation. Such new agents and therapeutic approaches will facilitate the development of effective treatment strategies and will ultimately improve the outcome for patients with this devastating disease. This article provides an overview of the approaches to the diagnosis, evaluation, and treatment of neoplastic meningitis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Meningeal Neoplasms/diagnosis , Meningitis/diagnosis , Cerebrospinal Fluid/cytology , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/therapeutic use , Genetic Therapy , Humans , Immunotherapy , Injections, Spinal , Meningeal Neoplasms/therapy , Meningitis/therapy , Prognosis
14.
Clin Cancer Res ; 6(3): 994-7, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741726

ABSTRACT

CI-994 is a substituted benzamide derivative that has demonstrated significant antitumor activity in vitro and in vivo against a broad spectrum of murine and human tumor models. Its mechanism of action is still unknown but seems to be novel compared with existing anticancer drugs. We studied the plasma and cerebrospinal fluid (CSF) pharmacokinetics of CI-994 in nonhuman primates. Three animals (total 4 doses) received an 80 mg/m2 dose of CI-994 administered over 20 min, and one animal received a dose of 100 mg/m2. Serial plasma and fourth ventricular CSF samples were obtained from 0 to 4320 min after administration of the 80-mg/m2 dose, and only plasma samples were obtained after the 100-mg/m2 dose. CI-994 was measured using a previously validated reverse-phase high-performance liquid chromatography assay. Elimination of CI-994 from plasma was triexponential (4 of 5 cases) or biexponential (1 of 5 cases), with a terminal half life (t1/2) of 7.4 +/- 2.5 h, volume of distribution of 15.5 +/- 1.8 L/m2, and clearance of 40 +/- 6 ml/min/m2. The area under the concentration-time curve (AUC) for the 80-mg/m2 dose was 125 +/- 17 microM x hr. CI-994 was first detected in CSF at the completion of the i.v. infusion. Peak concentrations of CI-994 in CSF were 3.4 +/- 0.3 microM. Elimination from CSF was monoexponential (2 of 4 cases) or biexponential (2 of 4 cases) with a terminal t1/2 in CSF of 12.9 +/- 2.5 h and AUC of 55 +/- 18 microM x hr. The AUC(CSF):AUCplasma ratio was 43 +/- 10%. This study demonstrates that there is excellent CSF penetration of CI-994 after i.v. administration. Additional studies are needed to evaluate the potential role of CI-994 in the treatment of central nervous system neoplasms.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Phenylenediamines/pharmacokinetics , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/cerebrospinal fluid , Area Under Curve , Benzamides , Infusions, Intravenous , Macaca mulatta , Male , Metabolic Clearance Rate , Phenylenediamines/blood , Phenylenediamines/cerebrospinal fluid
15.
Cancer Chemother Pharmacol ; 45(3): 259-64, 2000.
Article in English | MEDLINE | ID: mdl-10663645

ABSTRACT

PURPOSE: Intrathecal methotrexate achieves high concentrations in cerebrospinal fluid (CSF), but drug distribution throughout the subarachnoid space after an intralumbar dose is limited. The objective of this study was to quantify methotrexate distribution in CSF after intraventricular and intravenous administration and to identify factors that influence CSF distribution. METHODS: Nonhuman primates (Macaca mulatta) with permanently implanted catheters in the lateral and fourth ventricles received methotrexate by bolus injection (0.5 mg) and infusion (0.05 to 0.5 mg/day over 24 to 168 h) into the lateral ventricle, as well as intravenous infusions. CSF was sampled from the lumbar space, fourth ventricle and the subarachnoid space at the vertex. Methotrexate in CSF and plasma was measured with the dihydrofolate reductase inhibition assay. RESULTS: After bolus intraventricular injection, methotrexate exposure in lumbar CSF ranged from 11% to 69% of that achieved in the fourth ventricle. During continuous intraventricular infusions, methotrexate steady-state concentrations (C(ss)) in lumbar CSF and CSF from the vertex were only 20% to 25% of the ventricular CSF C(ss). The dose, duration of infusion, and infusate volume did not influence drug distribution to the lumbar CSF, but probenicid increased the lumbar to ventricular C(ss) ratio, suggesting the involvement of a probenicid-sensitive transport pump in the efflux of MTX from the CSF. During the intravenous infusions, the ventricular methotrexate C(ss) was lower than the lumbar C(ss) and the C(ss) in CSF from the vertex. CONCLUSION: Methotrexate CSF distribution after intraventricular injection was uneven, and at steady-state CSF methotrexate concentrations were lower at sites that were more distant from the injection site.


Subject(s)
Antimetabolites, Antineoplastic/cerebrospinal fluid , Methotrexate/cerebrospinal fluid , Subarachnoid Space/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacokinetics , Brain/drug effects , Brain/metabolism , Cerebral Ventricles/drug effects , Dose-Response Relationship, Drug , Infusions, Intravenous , Infusions, Parenteral , Injections, Intraventricular , Macaca mulatta , Male , Metabolic Clearance Rate , Methotrexate/pharmacokinetics , Probenecid/pharmacology , Uricosuric Agents/pharmacology
16.
J Pharmacol Exp Ther ; 291(2): 870-4, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10525111

ABSTRACT

Thiopurine antimetabolites have been in clinical use for more than 40 years, yet the metabolism of thiopurines remains only partially understood. Data from our previous pediatric phase 1 trial of continuous i.v. infusion of thioguanine (CIVI-TG) suggested that TG was eliminated by saturable mechanism, with conversion of the drug to an unknown metabolite. In this study we have identified this metabolite as 8-hydroxy-thioguanine (8-OH-TG). The metabolite coeluted with the 8-OH-TG standard on HPLC and had an identical UV spectrum, with a lambda(max) of 350 nm. On mass spectroscopy, the positive ion, single quad scan of 8-OH-TG yielded a protonated molecular ion at 184 Da and contained diagnostic ions at m/z 167, 156, 142, and 125 Da. Incubation of TG in vitro with partially purified aldehyde oxidase resulted in 8-OH-TG formation. 8-OH-TG is the predominant circulating metabolite found in patients receiving CIVI-TG and is likely generated by the action of aldehyde oxidase.


Subject(s)
Aldehyde Oxidoreductases/physiology , Antimetabolites, Antineoplastic/metabolism , Thioguanine/analogs & derivatives , Thioguanine/administration & dosage , Thioguanine/metabolism , Aldehyde Oxidase , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/blood , Child , Chromatography, High Pressure Liquid , Humans , In Vitro Techniques , Infusions, Intravenous , Mass Spectrometry , Thioguanine/blood
17.
Clin Cancer Res ; 5(4): 733-7, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10213206

ABSTRACT

Neutropenia is the dose-limiting toxicity of docetaxel in children. This Phase I trial was designed to determine the maximum tolerated dose, the dose-limiting toxicities, and the incidence and severity of other toxicities of docetaxel with filgrastim (G-CSF) support in children with refractory solid tumors. Docetaxel was administered as an i.v. infusion for 1 h every 21 days with a starting dose of 150 mg/m2 and an escalation to 185 mg/m2 and 235 mg/m2 in subsequent patient cohorts. G-CSF (5 microg/kg/day) was administered s.c., starting 48 h after docetaxel and continuing until the post-nadir neutrophil count reached 10,000/microl. Seventeen patients received 27 courses of docetaxel with G-CSF support. Generalized erythematous desquamating skin rash and myalgias were dose-limiting at 235 mg/m2. Localized and generalized rashes were seen at all of the three dose levels. Neutropenia (median nadir, 95/1microl) occurred at all of the dose levels but was brief in duration and not dose-limiting. Thrombocytopenia was minimal (median platelet count nadir, 139,000/microl), and the severity of neutropenia and thrombocytopenia did not seem to be related to the docetaxel dose. Other docetaxel-related toxicities included hemorrhage (associated with mucositis), sepsis, hypersensitivity reaction, transient elevation of liver enzymes, stomatitis, back pain, asthenia, and neuropathy. One minor response was observed in a patient with colon cancer. The maximum tolerated dose of docetaxel with G-CSF support in children is 185 mg/m2, which is 50% higher than the maximum tolerated dose of docetaxel alone in children and 85 % higher than the recommended adult dose.


Subject(s)
Granulocyte Colony-Stimulating Factor/administration & dosage , Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Taxoids , Adolescent , Adult , Child , Child, Preschool , Docetaxel , Dose-Response Relationship, Drug , Drug Eruptions , Female , Filgrastim , Humans , Infant , Male , Neutropenia/chemically induced , Neutropenia/prevention & control , Paclitaxel/administration & dosage , Paclitaxel/toxicity , Recombinant Proteins
18.
Cancer Chemother Pharmacol ; 43(5): 415-8, 1999.
Article in English | MEDLINE | ID: mdl-10100598

ABSTRACT

PURPOSE: The antiviral nucleoside analogue ganciclovir is a potent inhibitor of replication in herpes viruses and is effective against cytomegalovirus infections in immunocompromised patients. Ganciclovir is also used in cancer gene therapy studies that utilize the herpes simplex virus thymidine kinase gene (HSV-TK). The pharmacokinetics of ganciclovir in adults and children have been described previously but there are no detailed studies of the CNS pharmacology of ganciclovir. We studied the pharmacokinetics of ganciclovir in plasma and CSF in a nonhuman primate model that is highly predictive of the CSF penetration of drugs in humans. METHODS: Ganciclovir, 10 mg/kg i.v., was administered over 30 min to three animals. Ganciclovir concentrations in plasma and CSF were measured using reverse-phase HPLC. RESULTS: Peak plasma ganciclovir concentrations ranged from 18.3 to 20.0 microg/ml and the mean plasma AUC was 1075+/-202 microg/ml x min. Disappearance of ganciclovir from the plasma was biexponential with a distribution half-life (t(1/2)alpha) of 18+/-7 min and an elimination half-life (t(1/2)beta) of 109+/-7 min. Total body clearance (ClTB) was 9.4+/-1.6 ml/min/kg. The mean CSF ganciclovir AUC was 168+/-83 microg/ml x min and the mean peak CSF concentration was 0.7+/-0.3 microg/ml. The ratio of the AUCs in CSF and plasma was 15.5+/-7.1%. CONCLUSIONS: Ganciclovir penetrates into the CSF following i.v. administration. This finding will be useful in the design of gene therapy trials involving the HSV-TK gene followed by treatment with ganciclovir in CNS or leptomeningeal tumors.


Subject(s)
Antiviral Agents/blood , Antiviral Agents/cerebrospinal fluid , Ganciclovir/blood , Ganciclovir/cerebrospinal fluid , Animals , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Ganciclovir/adverse effects , Ganciclovir/pharmacokinetics , Macaca mulatta , Male , Metabolic Clearance Rate
20.
Clin Cancer Res ; 4(2): 357-60, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9516923

ABSTRACT

The antitumor activity of topotecan administered as a 72-h continuous i.v. infusion was evaluated in children with refractory neuroblastoma and sarcomas of soft tissue and bone. We also attempted to increase the dose intensity of topotecan by including an intrapatient dose escalation in the trial design. Ninety-three children (85 eligible and evaluable for response) with recurrent or refractory neuroblastoma, osteosarcoma, Ewing's sarcoma/peripheral neuroectodermal tumor, rhabdomyosarcoma, or other soft-tissue sarcomas received topotecan administered as a 72-h i.v. infusion every 21 days. The initial dose was 1.0 mg/m2/day, with subsequent intrapatient dose escalation to 1.3 mg/m2/day for those patients who did not experience dose-limiting toxicity after their first cycle of topotecan. There was one complete response in a patient with neuroblastoma (n = 26) and one partial response in a patient with Ewing's sarcoma/peripheral neuroectodermal tumor (n = 25). No complete or partial responses were observed in 17 patients with osteosarcoma, 15 patients with rhabdomyosarcoma, or 2 patients with other soft-tissue sarcomas; however, 8 patients had prolonged (15-48 weeks) stable disease while receiving topotecan. Topotecan was well tolerated. The most commonly observed toxicities were myelosuppression (dose-limiting) and nausea and vomiting. Intrapatient dose escalations were performed in 68% of the patients who received more than one cycle of topotecan, and 1.3 mg/m2/day was tolerated by 79% of the patients who received the higher dose and were evaluable for hematological toxicity. In conclusion, topotecan administered as a 72-h continuous infusion every 21 days is inactive (objective response rate, < 20%) in children with refractory or recurrent neuroblastoma and sarcomas of soft tissue or bone.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Topotecan/therapeutic use , Adolescent , Adult , Antineoplastic Agents/adverse effects , Bone Neoplasms/drug therapy , Child , Child, Preschool , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Infant , Infusions, Intravenous , Male , Neuroblastoma/drug therapy , Neuroectodermal Tumors, Primitive, Peripheral/drug therapy , Osteosarcoma/drug therapy , Rhabdomyosarcoma/drug therapy , Sarcoma, Ewing/drug therapy , Soft Tissue Neoplasms/drug therapy , Topotecan/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...