Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 245
Filter
2.
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
3.
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
4.
Drug Metab Dispos ; 27(10): 1128-32, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10497138

ABSTRACT

2'-beta-Fluoro-2',3'-dideoxyadenosine (F-ddA, lodenosine) is a nucleoside analog that was rationally designed as a more chemically and enzymatically stable anti-AIDS drug than its parent compound 2', 3'-dideoxyadenosine or didanosine. Plasma and cerebrospinal fluid (CSF) pharmacokinetics of this compound and its major metabolite, 2'-beta-fluoro-2',3'-dideoxyinosine (F-ddI), were studied in three rhesus monkeys after a single 20 mg/kg dose administered as an i.v. push. F-ddA exhibited a mean residence time of 0.17 h in plasma and its plasma concentration time profile appeared to be biexponential. The majority of plasma exposure was from F-ddI, with a mean parent drug area under the curve (AUC) to metabolite AUC ratio of 0.16. CSF levels were low, with a mean CSF AUC to plasma AUC ratio of 0.068, with approximately one-quarter of this exposure in CSF due to unchanged drug. Urinary excretion accounted for half of the drug administered with the majority recovered as the metabolite, F-ddI. In a separate experiment, one monkey received a 20 mg/kg i.v. dose of F-ddI. The total dideoxynucleoside plasma exposure was greater than it was after administration of F-ddA; however, the CSF AUC to plasma AUC ratio was a factor of 4 lower (0.017). Thus, F-ddA central nervous system penetration is at least comparable to that of didanosine, indicating that this experimental drug has potential as an addition to currently approved AIDS therapies.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Dideoxyadenosine/analogs & derivatives , Animals , Dideoxyadenosine/blood , Dideoxyadenosine/cerebrospinal fluid , Dideoxyadenosine/pharmacokinetics , Macaca mulatta , Male
6.
Cancer ; 85(2): 511-6, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-10023723

ABSTRACT

BACKGROUND: Intraventricular chemotherapy results in more uniform drug distribution within the subarachnoid space and allows for more flexible drug administration schedules. The authors report their experience with an intraventricular concentration times time (C x T) chemotherapy regimen for recurrent meningeal leukemia and lymphoma. METHODS: Twenty-one patients (median age, 11.6 years) received C x T therapy for meningeal acute lymphoblastic leukemia (n = 18), Burkitt's lymphoma (n = 2), or undifferentiated leukemia (n = 1). Prior therapy included standard intrathecal (IT) methotrexate and cytarabine, cranial or craniospinal radiation (median, 24 Gy), and 0-5 experimental treatment modalities. C x T induction therapy consisted of 2 mg of intraventricular methotrexate administered daily for 3 days every 10 days, for 4 courses. Patients were then consolidated with 4 courses of alternating intraventricular cytarabine (15 mg/day) or methotrexate (2 mg/day) daily for 3 days every 2 weeks (2 courses of methotrexate and 2 courses of cytarabine). Maintenance therapy consisted of alternating monthly courses of C x T methotrexate or cytarabine. RESULTS: Ninety-three percent of patients (14 of 15) who were evaluable for response achieved a complete remission in a median of 10 days (range, 2-40 days). Median remission duration was 15 months. Fourteen patients died of recurrent disease or systemic treatment-related complications; 2 patients are alive, off treatment, and in continuous complete remission for 59+ and 89+ months; 1 patient experienced a meningeal relapse at 24 months on C x T therapy but was reinduced with the C x T regimen, received craniospinal radiation, and is in remission at 142+ months; and 3 are alive with disease at 32+, 72+, and 81+ months. One patient was lost to follow-up. CONCLUSIONS: This regimen appears to be an effective and well-tolerated palliative treatment for patients with recurrent meningeal leukemia and lymphoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Burkitt Lymphoma/drug therapy , Meningeal Neoplasms/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Child, Preschool , Cytarabine/administration & dosage , Cytarabine/adverse effects , Female , Humans , Injections, Intraventricular , Male , Methotrexate/administration & dosage , Methotrexate/adverse effects , Recurrence , Remission Induction , Treatment Outcome
7.
J Pediatr Hematol Oncol ; 21(1): 26-30, 1999.
Article in English | MEDLINE | ID: mdl-10029808

ABSTRACT

PURPOSE: Idarubicin (4-demethoxy-daunorubicin) is more potent and less cardiotoxic than the commonly used anthracyclines, doxorubicin and daunorubicin. In addition, idarubicin is metabolized to an active metabolite, idarubicinol, in contrast to other anthracyclines whose alcohol metabolites are much less active than the parent drug. The current study was performed in nonhuman primates to determine the plasma and cerebrospinal fluid (CSF) pharmacokinetics of idarubicin and idarubicinol and to compare them to the pharmacokinetics of daunorubicin and daunorubicinol. METHODS: A dose of 30 mg/m2 of daunorubicin or 8 mg/m2 of idarubicin was administered intravenously over 15 minutes. Plasma and CSF were sampled frequently from the end of the infusion to 72 to 96 hours after infusion. Drug and metabolite concentrations were measured using high-pressure liquid chromatography (HPLC). RESULTS: Daunorubicin elimination from plasma was triphasic with a terminal half-life of 5.9 +/- 1.8 hours, area under the concentration-time curve (AUC) 22.5 +/- 9.2 mumol/L.min, and clearance 2790 +/- 960 mL/min/m2. Daunorubicinol elimination was biphasic with a terminal half-life 10.2 +/- 2.3 hours and an AUC 74.5 +/- 5.3 mumol/L.min. Idarubicin elimination was triphasic with terminal half-life of 12.3 +/- 11.4 hours, a AUC 10.8 +/- 3.7 mumol/L.min, and clearance 1650 +/- 610 mL/min/m2. Idarubicinol elimination was biphasic with a terminal half-life 28.7 +/- 4.2 hours and AUC 67 +/- 9.8 mumol/L.min. CSF penetration was low for both parent drugs and their metabolites. CSF idarubicin was measurable at a single time point (1 hour after administration) for 2 animals, and was not measurable for the third. The CSF to plasma concentration ratio at that time point was 8% in 1 animal and 15% in the other. Idarubicinol was detected in 2 to 4 samples at various times, appearing as early as 1 hour in 1 animal and persisting as late as 48 hours in another. The CSF to plasma concentration ratio at corresponding time points was 1.9 +/- 0.6%. Daunorubicin was measurable for < 6 hours after intravenous administration. For individual animals, the mean CSF to plasma concentration ranged from 4% to 12%. Daunorubicinol was detectable by 1 hour in 2 of 3 animals and by 3 hours in the other, and remained detectable at 24 hours in 2 of 3. The terminal half-life of daunorubicinol in CSF was 8.8 +/- 1.3 hours, the AUC was 1.8 +/- 1.5 mumol/L.min, and the AUCCSF to AUCplasma ratio was 2.4 +/- 1.9%. CONCLUSION: Idarubicin, idarubicinol, daunorubicin, and daunorubicinol penetrate poorly into the CSF after intravenous administration.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Daunorubicin/pharmacokinetics , Idarubicin/pharmacokinetics , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/blood , Antibiotics, Antineoplastic/cerebrospinal fluid , Chromatography, High Pressure Liquid , Daunorubicin/administration & dosage , Daunorubicin/blood , Daunorubicin/cerebrospinal fluid , Disease Models, Animal , Drug Administration Schedule , Idarubicin/administration & dosage , Idarubicin/blood , Idarubicin/cerebrospinal fluid , Infusions, Intravenous , Macaca mulatta , Male
8.
Blood ; 92(10): 3569-77, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9808549

ABSTRACT

We prospectively assessed the pharmacokinetics of methotrexate, mercaptopurine, and erythrocyte thioguanine nucleotide levels in a homogenous population of children with lower risk acute lymphoblastic leukemia and correlated pharmacokinetic parameters with disease outcome. The maintenance therapy regimen included daily oral mercaptopurine (75 mg/m2) and weekly oral methotrexate (20 mg/m2). One hundred ninety-one methotrexate doses and 190 mercaptopurine doses were monitored in 89 patients. Plasma drug concentrations of both agents were highly variable. The area under the plasma concentration-time curve (AUC) of methotrexate ranged from 0.63 to 12 micromol*h/L, and the AUC of mercaptopurine ranged from 0.11 to 8 micromol*h/L. Drug dose, patient age, and duration of therapy did not account for the variability. Methotrexate AUC was significantly higher in girls than boys (P =.007). There was considerable intrapatient variability for both agents. Erythrocyte thioguanine nucleotide levels were also highly variable (range, 0 to 10 pmol/g Hgb) and did not correlate with mercaptopurine dose or AUC. A Cox regression analysis showed that mercaptopurine AUC was a marginally significant (P =.043) predictor of outcome, but a direct comparison of mercaptopurine AUC in the remission and relapsed patient groups failed to show a significant difference. Methotrexate and mercaptopurine plasma concentrations and erythrocyte thioguanine nucleotide levels were highly variable, but measurement of these pharmacokinetic parameters at the start of maintenance will not distinguish patients who are more likely to relapse.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mercaptopurine/pharmacology , Methotrexate/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Administration, Oral , Adolescent , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/pharmacokinetics , Area Under Curve , Asparaginase/administration & dosage , Biological Availability , Child , Child, Preschool , Combined Modality Therapy , Cranial Irradiation , DNA Adducts , Erythrocytes/chemistry , Female , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/blood , Humans , Infant , Injections, Spinal , Male , Mercaptopurine/administration & dosage , Mercaptopurine/pharmacokinetics , Methotrexate/administration & dosage , Methotrexate/blood , Methotrexate/pharmacokinetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/radiotherapy , Prednisone/administration & dosage , Proportional Hazards Models , Recurrence , Thionucleotides/blood , Treatment Outcome , Vincristine/administration & dosage
10.
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
11.
Cancer Genet Cytogenet ; 101(2): 138-42, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9494616

ABSTRACT

A child with acute lymphoblastic leukemia (ALL) whose predominant leukemic clone demonstrated double minute chromosomes (dmin) is presented. The patient had no history of mutagen or carcinogen exposure and responded well to combination chemotherapy. Although dmin have been described in acute myelogenous leukemia and various solid tumors in adults, their presence in childhood neoplasms is less frequent and limited primarily to neurogenic tumors. This is the first documentation of dmin in childhood ALL, suggesting that there may be an unrecognized subgroup of ALL patients with gene amplification.


Subject(s)
Chromosome Aberrations , Gene Amplification , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Child , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male
12.
J Clin Oncol ; 16(1): 181-6, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9440741

ABSTRACT

PURPOSE: To define the maximum-tolerated dose (MTD), quantitative and qualitative toxicities, recommended phase II dose, and pharmacokinetics of pyrazoloacridine (PZA) administered as a 1- or 24-hour infusion in children and young adults with refractory cancers. PATIENTS AND METHODS: Twenty-two patients received PZA as a 1-hour infusion at doses of 380 mg/m2 (n = 3), 495 mg/m2 (n = 6), 640 mg/m2 (n = 6), and 835 mg/m2 (n = 7). An additional four patients received PZA as a 24-hour infusion at the MTD (640 mg/m2) for the 1-hour infusion schedule. Plasma samples were obtained for pharmacokinetic analysis in 17 patients. PZA concentration in plasma was measured by reverse-phase high-performance liquid chromatography (HPLC). A two-compartment pharmacokinetic model was fit to the PZA plasma concentration data. RESULTS: On the 1-hour infusion schedule, dose-limiting myelosuppression (neutropenia more than thrombocytopenia) was observed in two of seven patients at the 835-mg/m2 dose level. Myelosuppression did not appear to be ameliorated by prolonging the infusion to 24 hours. Nonhematologic toxicities were minor. Significant neurotoxicity, which was dose-limiting in adults treated with a 1-hour infusion of PZA, was observed in one patient treated at 640 mg/m2, but was not dose-limiting. There was marked interpatient variability in plasma PZA concentrations at all dose levels. The pharmacokinetic profile of PZA was characterized by an initial rapid decline (alpha half-life [t(1/2)alpha], 0.5 hours) followed by a prolonged elimination phase (t(1/2)beta, 30 hours). The volume of distribution at steady-state (Vd(ss)) was 700 L/m2 and the clearance was 300 mL/min/m2. There was no evidence of dose-dependent clearance. The area under the PZA concentration-time curve (AUC) correlated poorly with dose and was more predictive of the degree of myelosuppression than was PZA dose. CONCLUSION: PZA administered as 1- or 24-hour infusion is well tolerated by children and young adults. The dose-limiting toxicity (DLT) is myelosuppression. Neurotoxicity is not prominent in this age group. There was marked interpatient variation in plasma concentrations of PZA. The recommended dose for phase II studies is 640 mg/m2.


Subject(s)
Acridines/administration & dosage , Acridines/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy , Neoplasms/metabolism , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Acridines/adverse effects , Adolescent , Adult , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Child , Child, Preschool , Drug Administration Schedule , Female , Humans , Infant , Infusions, Intravenous , Male , Neutropenia/chemically induced , Pyrazoles/adverse effects , Pyrazoles/blood , Thrombocytopenia/chemically induced
13.
Int J Cancer Suppl ; 11: 48-51, 1998.
Article in English | MEDLINE | ID: mdl-9876478

ABSTRACT

It has been estimated that approximately 80% of the world's pediatric population lives in countries with limited resources, and that 43% of these children are malnourished. In children with cancer, malnutrition may antedate the diagnosis or be a result of aggressive chemotherapeutic regimens. Studies have shown that children with cancer and malnutrition have a less favorable prognosis, a higher risk of early relapse, and tolerate chemotherapy poorly when compared with children with normal nutritional status. Improvements in nutritional status may improve tolerance to chemotherapy. An understanding of the mechanisms responsible for the effects of malnutrition on drug disposition and pharmacodynamic response is important, especially for anti-neoplastic agents, which have a narrow therapeutic index and may be associated with potentially severe or life-threatening side-effects. Several factors related to malnutrition have been suggested to alter drug disposition. Diminished protein "status" in malnourished children results in lower amounts of plasma proteins, increasing the concentration of free drug available to exert its cytotoxic effect. Severely malnourished individuals also exhibit decreased oxidative metabolism and reduced glomerular filtration rate (GFR), potentially increasing concentrations of parent drug or active metabolites. Malnourished children receiving chemotherapy for the treatment of an underlying malignancy may need specifically "tailored" protocols to achieve therapeutic response while minimizing adverse acute and long-term side effects. The role of specific interventions, such as correction of nutritional status or pharmacokinetic drug monitoring, should be evaluated in this context.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Nutrition Disorders/metabolism , Nutritional Status , Absorption , Animals , Child , Glomerular Filtration Rate , Humans , Liver/metabolism , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/metabolism , Nutrition Disorders/complications , Protein Binding
14.
Clin Cancer Res ; 3(5): 713-7, 1997 May.
Article in English | MEDLINE | ID: mdl-9815740

ABSTRACT

Although mercaptopurine is the thiopurine antimetabolite predominantly used in the treatment of childhood acute lymphoblastic leukemia (ALL), thioguanine (TG) is more potent than mercaptopurine in in vitro cytotoxicity studies in human leukemic cell lines and leukemic cells from patients with ALL. We conducted a pediatric Phase I trial of TG administered as a continuous i.v. infusion (CIV). A pharmacokinetically guided dose escalation was performed to define the dose rate of TG required to achieve a steady-state plasma concentration (Css) exceeding the target concentration of 1 microM, and then the maximum tolerated duration of infusion of TG at this dose rate was defined. Eighteen patients (median age, 18 years; range, 4-25 years) with refractory malignancies (16 solid tumors and 2 ALL) were enrolled in this study. The starting dose rate of 10 mg/m2/h administered for 24 h achieved an average Css of 0.9 microM (range, 0.7-1.2 microM). Therefore, the dose rate was escalated to 20 mg/m2/h, which achieved an average Css of 4.1 microM (range, 1. 0-8.3 microM). This disproportionate increase in the Css of TG suggested a capacity-limited (saturable) elimination process, and a pharmacokinetic model incorporating two compartments with capacity-limited elimination from the central compartment was developed to describe the disposition of TG. The TG clearances (derived from model parameters) at the 10- and 20-mg/m2/h dose rates were 987 and 608 ml/min/m2, respectively. Dose-limiting myelosuppression (absolute granulocyte count < 500/mm3 and platelet count < 25,000/mm3) was observed in two of three patients treated with a dose rate of 20 mg/m2/h administered for 36 h. Administration of CIV of TG at 20 mg/m2/h for 24 h was well tolerated in nine patients. Nonhematological toxicities included nonneutropenic infections and mild, reversible changes in hepatic function tests. The recommended dose rate and duration for CIV of TG is 20 mg/m2/h for 24 h.


Subject(s)
Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/pharmacokinetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Thioguanine/adverse effects , Thioguanine/pharmacokinetics , Adolescent , Adult , Antimetabolites, Antineoplastic/administration & dosage , Child , Child, Preschool , Female , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Models, Biological , Thioguanine/administration & dosage
15.
J Clin Oncol ; 15(4): 1538-43, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9193350

ABSTRACT

PURPOSE: A phase I trial of docetaxel was performed to determine the maximum-tolerated dose (MTD), the dose-limiting toxicities, and the incidence and severity of other toxicities in children with refractory solid tumors. PATIENTS AND METHODS: Forty-four children received 103 courses of docetaxel administered as a 1-hour intravenous infusion every 21 days. Doses ranged from 55 to 150 mg/m2, MTD was defined in heavily pretreated and less heavily pretreated (< or = 2 prior chemotherapy regimens, no prior bone marrow transplantation [BMT], and no radiation to the spine, skull, ribs, or pelvic bones) patients. RESULTS: Dose-related neutropenia was the primary dose-limiting toxicity. The MTD in the heavily pretreated patient group was 65 mg/m2, but the less heavily pretreated patients tolerated a significantly higher dose of docetaxel (maximum-tolerated dose, 125 mg/m2). Neutropenia and constitutional symptoms consisting of malaise, myalgias, and anorexia were the dose-limiting toxicities at 150 mg/m2 in the less heavily pretreated patients. Thrombocytopenia was not prominent, even in patients who experienced dose-limiting neutropenia. Common nonhematologic toxicities of docetaxel included skin rashes, mucositis, and mild elevations of serum transaminases. Neuropathy was uncommon. Peripheral edema and weight gain were observed in two of five patients who received more than three cycles of docetaxel. A complete response (CR) was observed in one patient with rhabdomyosarcoma, a partial response (PR) in one patient with peripheral primitive neuroectodermal tumor (PPNET), and a minimal response (MR) in two patients with PPNET. Three of the four responding patients were treated at doses > or = 100 mg/m2. CONCLUSION: The recommended phase II dose of docetaxel administered as a 1-hour intravenous infusion in children with solid tumors in 125 mg/m2. Because neutropenia was the dose-limiting toxicity and thrombocytopenia was mild, further escalation of the dose should be attempted with granulocyte colony-stimulating factor (G-CSF) support.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Taxoids , Adolescent , Adult , Antineoplastic Agents, Phytogenic/adverse effects , Child , Child, Preschool , Docetaxel , Drug Administration Schedule , Female , Humans , Incidence , Infant , Infusions, Intravenous , Male , Neutropenia/chemically induced , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Severity of Illness Index , Treatment Outcome
16.
Cancer ; 78(3): 527-31, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8697400

ABSTRACT

BACKGROUND: Topotecan is a topoisomerase I inhibitor that has good penetration across the blood-brain barrier and significant antitumor activity against human brain tumor xenografts. In a Phase I trial in children with refractory cancer, topotecan was well tolerated when administered as a 24-hour infusion. The maximum tolerated dose was 5.5 mg/m2 and the dose-limiting toxicity was myelosuppression. This Phase II study of topotecan was performed to assess the activity of topotecan against childhood brain tumors. METHODS: Forty-five children with either a previously treated primary brain tumor that was refractory to standard therapy, or an untreated brain stem glioma or glioblastoma multiforme, received topotecan administered as a 24-hour intravenous infusion every 21 days. The initial dose was 5.5 mg/m2 with escalation to 7.5 mg/m2 on the second and subsequent doses in patients who did not experience dose-limiting toxicity. RESULTS: There were no complete or partial responses in the patients with high grade glioma (n=9), medulloblastoma (n=9), or brain stem glioma (n=14). One of 2 patients with a low grade glioma had a partial response lasting more than 17 months; 3 patients with a brain stem glioma had stable disease for 12 to 28 weeks; and 1 patient with a malignant neuroepithelial tumor and 1 patient with an optic glioma had stable disease for 41 weeks and 22 weeks, respectively. Dose escalation from 5.5 mg/m2 to 7.5 mg/m2 was well tolerated in the first 11 patients enrolled on this study who had not received prior craniospinal radiation therapy. The starting dose was subsequently increased to 7.5 mg/m2 for patients without prior craniospinal radiation. CONCLUSIONS: Topotecan administered as a 24-hour infusion every 21 days is inactive in high grade gliomas, medulloblastomas, and brain stem tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Camptothecin/analogs & derivatives , Adolescent , Adult , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Brain Stem , Camptothecin/administration & dosage , Camptothecin/adverse effects , Camptothecin/therapeutic use , Child , Child, Preschool , Drug Administration Schedule , Female , Glioblastoma/drug therapy , Glioma/drug therapy , Humans , Infant , Infusions, Intravenous , Male , Topotecan
17.
J Clin Oncol ; 14(3): 774-82, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8622023

ABSTRACT

PURPOSE: We conducted a phase I/II trial of concurrently administered 72-hour infusional paclitaxel and doxorubicin in combination with granulocyte colony-stimulating factor (G-CSF) in patients with previously untreated metastatic breast cancer and bidimensionally measurable disease. PATIENTS AND METHODS: We defined the maximum-tolerated dose (MTD) of concurrent paclitaxel and doxorubicin administration and then studied potential pharmacokinetic interactions between the two drugs. Forty-two patients who had not received prior chemotherapy for metastatic breast cancer received 296 total cycles of paclitaxel and doxorubicin with G-CSF. RESULTS: The MTD was determined to be paclitaxel 180 mg/m2 and doxorubicin 60 mg/m2 each by 72-hour infusion with G-CSF. Diarrhea was the dose-limiting toxicity (DLT) of this combination, with three of three patients developing abdominal computed tomographic (CT) scan evidence of typhlitis (cecal thickening) at the dose level above the MTD. All patients developed grade 4 neutropenia (absolute neutrophil count [ANC] < 500 microL), generally less than 5 days in duration. This combination was generally safely administered at dose levels at or below the MTD. The overall response rate was 72% (28 of 39 patients; 95% confidence interval [CI], 55% to 85%), with 8% complete responses (CRs) (three of 39; 95% CI, 2% to 21%) and a median response duration of 9 months. The median overall survival time for all patients is 23 months, with a median follow-up duration of 28 months. Pharmacokinetic studies showed that administration of paclitaxel and doxorubicin together by 72-hour infusion did not affect the steady-state concentrations of either drug. CONCLUSION: Concurrent 72-hour infusional paclitaxel and doxorubicin can be administered safely, but is associated with significant toxicity. The overall response rate of this combination in untreated metastatic breast cancer patients is similar to that achieved with other doxorubicin-based combination regimens. The modest complete response rate achieved suggests that this schedule of paclitaxel and doxorubicin administration does not produce significant additive or synergistic cytotoxicity against breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Adult , Aged , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Diarrhea/chemically induced , Disease-Free Survival , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Doxorubicin/pharmacokinetics , Drug Administration Schedule , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Heart/drug effects , Humans , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Paclitaxel/pharmacokinetics , Thrombocytopenia/chemically induced
18.
J Clin Oncol ; 14(2): 362-72, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8636745

ABSTRACT

PURPOSE: We conducted an open-label, randomized trial to determine whether ICRF-187 would reduce doxorubicin-induced cardiotoxicity in pediatric sarcoma patients. METHODS: Thirty-eight patients were randomized to receive doxorubicin-containing chemotherapy (given as an intravenous bolus) with or without ICRF-187. Resting left ventricular ejection fraction (LVEF) was monitored serially with multigated radionuclide angiography (MUGA) scan. The two groups were compared for incidence and degree of cardiotoxicity, response rates to four cycles of chemotherapy, event-free and overall survival, and incidence and severity of noncardiac toxicities. RESULTS: Eighteen ICRF-187-treated and 15 control patients were assessable for cardiac toxicity. ICRF-187-treated patients were less likely to develop subclinical cardiotoxicity (22% v 67%, P < .01), had a smaller decline in LVEF per 100 mg/m2 of doxorubicin (1.0 v 2.7 percentage points, P = .02), and received a higher median cumulative dose of doxorubicin (410 v 310 mg/m2, P < .05) than did control patients. Objective response rates were identical in the two groups, with no significant differences seen in event-free or overall survival. ICRF-187-treated patients had a significantly higher incidence of transient grade 1 serum transaminase elevations and a trend toward increased hematologic toxicity. CONCLUSION: ICRF-187 reduces the risk of developing short-term subclinical cardiotoxicity in pediatric sarcoma patients who receive up to 410 mg/m2 of doxorubicin. Response rates to chemotherapy, event-free and overall survival, and noncardiac toxicities appear to be unaffected by the use of ICRF-187. Additional clinical trials with larger numbers of patients are needed to determine if the short-term cardioprotection afforded by ICRF-187 will reduce the incidence of late cardiac complications in long-term survivors of childhood cancer.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Cardiovascular Agents/therapeutic use , Doxorubicin/adverse effects , Heart/drug effects , Razoxane/therapeutic use , Sarcoma/drug therapy , Soft Tissue Neoplasms/drug therapy , Adolescent , Adult , Cardiovascular Agents/pharmacokinetics , Child , Female , Humans , Injections, Intravenous , Male , Neuroectodermal Tumors, Primitive, Peripheral/drug therapy , Razoxane/pharmacokinetics , Rhabdomyosarcoma/drug therapy , Sarcoma/mortality , Sarcoma, Ewing/drug therapy , Stroke Volume/drug effects , Survival Rate , Transaminases/blood
19.
Invest New Drugs ; 13(4): 337-42, 1996.
Article in English | MEDLINE | ID: mdl-8824353

ABSTRACT

A Phase II study of thioTEPA was performed by the Children's Cancer Group. ThioTEPA was administered intravenously every three weeks, at a dose of 65 mg/m2. Pediatric patients with recurrent sarcomas were targeted, but patients with other tumor diagnoses were also eligible. Toxicity was primarily hematopoietic, with thrombocytopenia being predominant. ThioTEPA did not demonstrate significant activity in the target tumor groups evaluated.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Neoplasms/drug therapy , Thiotepa/therapeutic use , Adolescent , Bone Neoplasms/drug therapy , Child , Humans , Neuroectodermal Tumors, Primitive, Peripheral/drug therapy , Osteosarcoma/drug therapy , Rhabdomyosarcoma/drug therapy , Sarcoma/drug therapy , Sarcoma, Ewing/drug therapy
20.
Curr Opin Oncol ; 8(1): 13-9, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8868094

ABSTRACT

The treatment of central nervous system (CNS) leukemia still poses a significant challenge to the clinical oncologist despite significant advances in therapeutic strategies that are directly targeted at the CNS. This article reviews the evolving definition of CNS leukemia, the current status of therapy for the prevention and treatment of overt CNS leukemia, pharmacological considerations in the treatment of CNS leukemia, and the longterm sequelae of CNS-directed therapy, focusing on articles that have been published in the past 2 years. New agents and treatment strategies for the treatment and prevention of CNS leukemia are also discussed.


Subject(s)
Central Nervous System Diseases/therapy , Leukemia/therapy , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Combined Modality Therapy , Genetic Therapy , Humans , Injections, Spinal , Leukemia/diagnosis , Meningeal Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...