Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Leuk Lymphoma ; 43(10): 1945-50, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12481889

ABSTRACT

To determine the maximum tolerated dose (MTD) and assess the toxicity profile and pharmacokinetics of weekly gemcitabine infusions in pediatric patients with refractory hematologic malignancies. Fourteen patients under 21 years old were given infusions of gemcitabine for escalating durations at 10 mg/m2/min weekly for three consecutive weeks. Two males and two females were studied at each dose level. Pharmacokinetics of the drug's metabolism were measured by high pressure-liquid chromatography (HPLC) for 24 h after the first dose. Intracellular difluorodeoxycytidine triphosphate formation in leukemic blasts was measured in selected patients. The MTD of gemcitabine in these patients was 3600 mg/m2/week for three consecutive weeks (10 mg/m2/min for 360 min). Hepatotoxicity was the dose limiting toxicity. Thirty to fifty percent of patients exhibited fever, rash, or myalgia. Rare instances of hypotension and pulmonary toxicity were observed. Two of six patients [one acute lymphoblastic leukemia (ALL) and one acute myelogenous leukemia (AML)] treated at the MTD had at least M2 marrows, although peripheral blood counts did not recover sufficiently for the patients to be considered in complete response. Pharmacokinetics of gemcitabine fit a two-compartment open model with terminal half-life and plasma clearance value of 62 min and 2.2 l/min/m2, respectively. No gender differences were observed. In conclusion, the MTD of gemcitabine was 10 mg/m2/min for 360 min every week for 3 weeks. This is the recommended phase II dose schedule for children with leukemia. The activity of the drug at this schedule in heavily pretreated, refractory patients warrants a phase II trial in hematologic malignancies.


Subject(s)
Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Leukemia/drug therapy , Salvage Therapy/methods , Adolescent , Biotransformation , Chemical and Drug Induced Liver Injury , Child , Child, Preschool , Chromatography, High Pressure Liquid , Deoxycytidine/pharmacokinetics , Deoxycytidine/toxicity , Female , Half-Life , Humans , Infant , Leukemia/complications , Male , Maximum Tolerated Dose , Metabolic Clearance Rate , Gemcitabine
2.
Clin Cancer Res ; 7(10): 3034-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11595692

ABSTRACT

PURPOSE: To determine the maximum tolerated dose and describe the toxicities of 9-cis-retinoic acid (9cRA, ALRT1057) administered p.o. tid in pediatric patients with refractory cancer and to study the pharmacokinetics of 9cRA and determine whether systemic drug exposure changes with chronic dosing. PATIENTS AND METHODS: Children with refractory cancer (stratified by age, < or =12 and >12 years) were treated with p.o. 9cRA for 28 consecutive days. The starting dose was 50 mg/m(2)/day divided into 3 doses with planned escalations to 65, 85, and 110 mg/m(2)/day. Pharmacokinetic sampling was performed on days 1 and 29 of the first cycle. RESULTS: Of the 37 patients entered, 18 patients < or =12 years of age and 11 patients >12 years of age were evaluable for toxicity. In patients >12 years of age, dose-limiting headache occurred in 2/2 patients at the 110 mg/m(2)/day dose level; 1/8 patients at 85 mg/m(2)/day developed dose-limiting pseudotumor cerebri. In patients < or =12 years of age, 3/5 patients at the starting dose level of 50 mg/m(2)/day developed dose-limiting pseudotumor cerebri; and 0/6 patients experienced dose-limiting toxicity at 35 mg/m(2)/day. Reversible non-dose-limiting hepatotoxicity was observed in 15 patients across all of the dose levels. There was considerable interpatient variability in 9cRA plasma concentrations. Peak plasma concentrations of 9cRA occurred at a median of 1.5 h after a p.o. dose, and the harmonic-mean terminal half-life was 43 min. By day 29 of 9cRA administration, the plasma 9cRA area under the curve declined by an average of 65% from day 1 values. CONCLUSIONS: The dose-limiting toxicity of 9cRA in pediatric patients was neurotoxicity, primarily pseudotumor cerebri. Younger children tolerate significantly lower doses of 9cRA than older children. Similar to all-trans-retinoic acid, the pharmacokinetics of 9cRA demonstrated a wide degree of interpatient variability and decreased over time when administered on a daily basis. The recommended Phase II dose of 9cRA in patients < or =12 and >12 years of age is 35 and 85 mg/m(2)/day, respectively.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Tretinoin/therapeutic use , Adolescent , Adult , Age Factors , Alitretinoin , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Headache/chemically induced , Humans , Liver/enzymology , Male , Nausea/chemically induced , Neoplasms/metabolism , Skin Diseases/chemically induced , Transaminases/drug effects , Transaminases/metabolism , Treatment Outcome , Tretinoin/adverse effects , Tretinoin/pharmacokinetics , Triglycerides/blood , Vomiting/chemically induced
3.
Pediatr Infect Dis J ; 18(8): 702-8, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10462340

ABSTRACT

BACKGROUND: Lipid formulations of amphotericin B have been recently introduced for treatment of invasive fungal infections. However, little is known about their role in pediatric populations. METHODS: We studied the safety and antifungal efficacy of amphotericin B lipid complex (ABLC, Abelcet) in 111 treatment episodes in pediatric patients through an open label, emergency use multicenter study. Patients with invasive fungal infections were enrolled if they had mycoses refractory to conventional antifungal therapy, if they were intolerant of previous systemic antifungal agents or concomitant nephrotoxic drugs or if they had preexisting renal disease. RESULTS: All 111 treatment episodes were evaluable for safety and 54 were evaluable for efficacy. The mean serum creatinine for the study population did not significantly change between baseline (1.23 +/- 0.11 mg/dl) and cessation of ABLC therapy (1.32 +/- 0.12 mg/dl) during 6 weeks. There were no significant differences observed between initial and end-of-therapy levels of serum potassium, magnesium, aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase and hemoglobin. However, there was an increase in mean total bilirubin (3.66 +/- 0.73 to 5.31 +/- 1.09 mg/dl) at the end of therapy (P = 0.054). Among 54 cases fulfilling criteria for evaluation of antifungal efficacy, a complete or partial therapeutic response was obtained in 38 patients (70%) after ABLC therapy. Complete or partial therapeutic response was documented in 56% of cases with aspergillosis (n = 25) and in 81% (n = 27) with candidiasis. Among premature infants (n = 8) and allogeneic marrow recipients (n = 14), response rates were 88 and 57%, respectively. Response was similar in those patients enrolled because of intolerance to previous antifungal therapy or because of progressive infection. CONCLUSIONS: These data support the use of ABLC for treatment of invasive fungal infections in pediatric patients who are intolerant of or refractory to conventional antifungal therapy.


Subject(s)
Amphotericin B/therapeutic use , Antifungal Agents/therapeutic use , Mycoses/drug therapy , Phosphatidylcholines/therapeutic use , Phosphatidylglycerols/therapeutic use , Adolescent , Amphotericin B/administration & dosage , Amphotericin B/adverse effects , Antifungal Agents/administration & dosage , Antifungal Agents/adverse effects , Aspergillosis/drug therapy , Candidiasis/drug therapy , Child , Child, Preschool , Creatinine/blood , Drug Combinations , Female , Humans , Infant , Infant, Newborn , Male , Phosphatidylcholines/administration & dosage , Phosphatidylcholines/adverse effects , Phosphatidylglycerols/administration & dosage , Phosphatidylglycerols/adverse effects , Zygomycosis/drug therapy
4.
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
5.
Clin Cancer Res ; 5(12): 3956-62, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10632325

ABSTRACT

The purpose of this study was to determine the toxicity, maximum tolerated dose, and pharmacokinetics of a 21-day continuous infusion of topotecan in children with relapsed solid tumors. Fifteen patients received 40 courses of continuous ambulatory infusions of topotecan every 28 days or when there was resolution of hematological toxicity and any grade 2 or greater nonhematological toxicity. The starting dose was 0.4 mg/m2/day. Total topotecan levels were measured on days 1, 7, 14, and 21. Three of four patients who received a starting dose of 0.4 mg/m2/day experienced dose-limiting myelosuppression. At the reduced dose of 0.3 mg/ m2/day, only two of the seven patients experienced dose-limiting myelosuppression. Subsequently, four patients with more limited prior therapy were treated with 0.4 mg/m2/ day; three had dose-limiting myelosuppression. Two patients with a dose-limiting toxicity at 0.4 mg/m2/day tolerated additional courses at 0.3 mg/m2/day. An equal number of patients had grade 4 neutropenia or thrombocytopenia. Other adverse events were rare. Two patients with ependymoma, one with rhabdomyosarcoma, and one with retinoblastoma metastatic to the brain had objective responses. The steady state plasma concentration and clearance of topotecan (Css) was achieved by day 1. Css in six patients with complete data were 1.44 +/- 0.50 and 2.13 +/- 0.83 ng/ml at 0.3 and 0.4 mg/m2/day, respectively. Thus, a 21-day topotecan infusion was well-tolerated at 0.3 mg/m2/day. Myelo-suppression was the dose-limiting toxicity at 0.4 mg/m2/day. The steady state and clearance of topotecan in this study are similar to those reported in adult patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Neoplasm Recurrence, Local/drug therapy , Topotecan/administration & dosage , Topotecan/adverse effects , Adolescent , Adult , Antineoplastic Agents/pharmacokinetics , Child , Child, Preschool , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Infusions, Intravenous , Male , Neoplasm Recurrence, Local/blood , Topotecan/pharmacokinetics
6.
J Clin Oncol ; 16(9): 3037-43, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9738573

ABSTRACT

PURPOSE: The Children's Cancer Group conducted a phase I trial of temozolomide stratified by prior craniospinal irradiation (CSI). PATIENTS AND METHODS: Children and adolescents with recurrent or progressive cancer were enrolled. Temozolomide was administered orally daily for 5 days, with subsequent courses administered every 21 to 28 days after full hematologic recovery. Dose levels tested included 100, 150, 180, 215, 245, and 260 mg/m2 daily. RESULTS: Twenty-seven patients on the non-CSI stratum were assessable for hematologic toxicity. During the first three dose levels (100, 150, and 180 mg/m2 daily), only grades 1 and 2 hematologic toxicity occurred. One patient at 215 mg/m2 daily had grade 3 hematologic toxicity. Three of eight patients (38%) treated at 245 to 260 mg/m2 daily had dose-limiting toxicity (DLT), which included both neutropenia and thrombocytopenia. Twenty-two patients on the CSI stratum were assessable for hematologic toxicity. Hematologic DLT occurred in one of six patients (17%) at 100 mg/m2 daily and in two of four patients (50%) at 215 mg/m2 daily. No nonhematologic DLT occurred; nausea and vomiting occurred in more than half of the patients. After two courses of temozolomide, 10 patients had stable disease (SD), and three patients had a partial response (PR), one of whom subsequently had a complete response (CR) that persists through 24 months of follow-up. CONCLUSION: The maximum-tolerated dose (MTD) of temozolomide for children and adolescents without prior CSI is 215 mg/m2 daily and for those with prior CSI is 180 mg/m2 daily for 5 days, with subsequent courses that begin on day 28. Temozolomide is well tolerated and should undergo phase II testing in children and adolescents.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/analogs & derivatives , Neoplasm Recurrence, Local/drug therapy , Neoplasms/drug therapy , Adolescent , Adult , Antineoplastic Agents, Alkylating/adverse effects , Child , Child, Preschool , Dacarbazine/adverse effects , Dacarbazine/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Male , Temozolomide
7.
Clin Infect Dis ; 26(6): 1383-96, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9636868

ABSTRACT

The safety and antifungal efficacy of amphotericin B lipid complex (ABLC) were evaluated in 556 cases of invasive fungal infection treated through an open-label, single-patient, emergency-use study of patients who were refractory to or intolerant of conventional antifungal therapy. All 556 treatment episodes were evaluable for safety. During the course of ABLC therapy, serum creatinine levels significantly decreased from baseline (P < .02). Among 162 patients with serum creatinine values > or = 2.5 mg/dL at the start of ABLC therapy (baseline), the mean serum creatinine value decreased significantly from the first week through the sixth week (P < or = .0003). Among the 291 mycologically confirmed cases evaluable for therapeutic response, there was a complete or partial response to ABLC in 167 (57%), including 42% (55) of 130 cases of aspergillosis, 67% (28) of 42 cases of disseminated candidiasis, 71% (17) of 24 cases of zygomycosis, and 82% (9) of 11 cases of fusariosis. Response rates varied according to the pattern of invasive fungal infection, underlying condition, and reason for enrollment (intolerance versus progressive infection). These findings support the use of ABLC in the treatment of invasive fungal infections in patients who are intolerant of or refractory to conventional antifungal therapy.


Subject(s)
Amphotericin B/therapeutic use , Antifungal Agents/therapeutic use , Mycoses/drug therapy , Phosphatidylcholines/therapeutic use , Phosphatidylglycerols/therapeutic use , Adult , Amphotericin B/adverse effects , Antifungal Agents/adverse effects , Aspergillosis/drug therapy , Candidiasis/drug therapy , Creatinine/blood , Cryptococcosis/drug therapy , Drug Combinations , Female , Humans , Male , Phosphatidylcholines/adverse effects , Phosphatidylglycerols/adverse effects
8.
Haemostasis ; 28(2): 93-8, 1998.
Article in English | MEDLINE | ID: mdl-10087434

ABSTRACT

Activated recombinant human coagulation factor VII (rFVIIa) is a promising new therapeutic agent for patients with hemophilia A or B with inhibitors who experience serious bleeding episodes or who need coverage during surgical procedures. This open-label, uncontrolled, emergency-use study evaluated the efficacy and safety of rFVIIa in 11 hemophiliac patients and 1 FVII-deficient patient with life-threatening intracranial hemorrhage previously unresponsive to one or more alternative therapies. rFVIIa effectively controlled intracranial hemorrhage in 10 of the 12 patients. Patients with hemophilia A or B received an average of 96.9 rFVIIa injections over 14.7 days with a mean total administration of 153.3 mg, corresponding to 8.1 mg/kg. Most reported adverse events were considered to be unrelated to rFVIIa therapy. These findings suggest that rFVIIa is an effective and well-tolerated therapeutic option in the management of central nervous system bleeding in patients with hemophilia A or B with inhibitors.


Subject(s)
Cerebral Hemorrhage/drug therapy , Factor VIIa/administration & dosage , Hemophilia A/complications , Hemophilia B/complications , Adult , Cerebral Hemorrhage/etiology , Child, Preschool , Factor VIIa/adverse effects , Humans , Infant , Injections, Intravenous , Male , Recombinant Proteins/administration & dosage
9.
J Clin Oncol ; 15(8): 2780-5, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9256119

ABSTRACT

PURPOSE: The Children's Cancer Group (CCG) undertook a phase I study (CCG-0922) to determine a tolerable dose of idarubicin given with fludarabine and cytarabine in children with relapsed or refractory leukemia. The phase I study was extended to a limited phase II study to assess the activity of this combination in children with acute myelogenous leukemia (AML). PATIENTS AND METHODS: This was a multiinstitutional study within the CCG. Eleven patients were entered onto the phase I study: seven with AML, three with acute lymphoblastic leukemia (ALL), and one with chronic myelogenous leukemia (CML). The maximal-tolerated dose (MTD) of fludarabine and cytarabine determined in a previous study was a fludarabine loading dose (LD) of 10.5 mg/m2 followed by a continuous infusion (CI) of 30.5 mg/m2/24 hours for 48 hours, followed by cytarabine LD 390 mg/m2, then CI 101 mg/m2/h for 72 hours. Idarubicin was given at three dose levels: 6, 9, and 12 mg/m2 intravenously (I.V.) on days 0, 1, and 2. The phase II portion of the trial included 10 additional patients with relapsed or refractory AML. RESULTS: A dose of idarubicin 12 mg/m2/d for 3 days given in combination with fludarabine and cytarabine was tolerated. The major toxicity encountered was hematologic. Nonhematologic toxicities included transaminase elevations, hyperbilirubinemia, and infections. Eight of 10 patients with AML in the phase II portion (12 mg/m2 idarubicin) achieved a complete remission (CR). CONCLUSION: This combination is active in patients with relapsed or refractory AML. The major toxicity encountered is hematologic. This regimen may be useful therapy for AML and should be compared with standard induction therapy in children with newly diagnosed AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia/drug therapy , Acute Disease , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Child, Preschool , Cytarabine/administration & dosage , Cytarabine/adverse effects , Female , Humans , Idarubicin/administration & dosage , Idarubicin/adverse effects , Infant , Infusions, Intravenous , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Vidarabine/administration & dosage , Vidarabine/adverse effects , Vidarabine/analogs & derivatives
10.
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
12.
Ann Surg Oncol ; 3(6): 539-42, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8915485

ABSTRACT

BACKGROUND: Video-assisted thoracic surgery (VATS) may complement open thoracotomy in children with osteosarcoma requiring pulmonary metastasectomy. METHODS: The records of children with metastatic pulmonary osteosarcoma considered for initial VATS intervention (n = 9) were reviewed. RESULTS: Two children did not have VATS exploration: one child with multiple bilateral nodules and another child with a deep parenchymal nodule. VATS provided diagnostic biopsy material in all cases when used (n = 7). Two children had benign inflammatory lesions; four children had VATS-directed wedge resections of solitary malignant lesions; and one child had VATS biopsy of diffuse parenchymal and pleural pulmonary disease not amenable to resection. The mean operative time and hospital length of stay were 1.78 +/- 0.54 h and 3.5 +/- 1.8 days, respectively. There were two complications of VATS: bleeding in a child, requiring a transfusion, and a latent pneumothorax in a patient after removal of the chest tube. CONCLUSION: VATS is safe, serves as an excellent diagnostic modality, complements the open thoracotomy, and may enable the surgeon to avoid more extensive procedures in selected cases.


Subject(s)
Lung Neoplasms/secondary , Lung Neoplasms/surgery , Osteosarcoma/secondary , Osteosarcoma/surgery , Thoracotomy/methods , Adolescent , Adult , Biopsy , Child , Disease-Free Survival , Female , Humans , Length of Stay , Lung Neoplasms/pathology , Male , Osteosarcoma/pathology , Postoperative Complications , Time Factors , Video Recording
13.
J Clin Oncol ; 14(3): 935-40, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8622042

ABSTRACT

PURPOSE: We have observed a severe atypical neuropathy (SAN) in patients with small non-cleaved-cell (SNCL) and large-cell lymphoma (LCL) treated with intensive chemotherapy and hematopoietic colony-stimulating factors (CSFs). The present analysis was undertaken in an attempt to identify factors associated with the development of this syndrome. PATIENTS AND METHODS: Fifty-four adult and pediatric patients consecutively treated according to the same chemotherapy protocol were included in the analysis. Low-risk patients received three cycles of cyclophosphamide, vincristine, doxorubicin, and high-dose methotrexate (CODOX-M) while in high-risk patients this drug combination was alternated with high-dose cytarabine (ara-C), etoposide, and ifosfamide (IVAC) for a total of four cycles. Twenty-eight patients received a CSF (granulocyte [G]- or granulocyte-macrophage [GM]-CSF), and 26 patients received no CSF. A statistical analysis, which included a logistic regression model, was undertaken to examine the importance of potential contributing factors to the development of SAN. RESULTS: SAN, which consisted of excruciating foot pain, usually associated with marked motor weakness, was observed in 12 patients. There was a highly significant association between the occurrence of this syndrome and the administration of CSFs, and an independent association with the cumulative dose of vincristine given in the first cycle of chemotherapy. Furthermore, the analysis suggested a synergistic effect between administration of the CSFs and vincristine in the genesis of this neuropathy. CONCLUSION: Our results indicate that CSFs can precipitate SAN when given in conjunction with vincristine. The development of SAN was associated most strongly with the cumulative dose of vincristine -- the size of individual doses and the number of doses given in cycle 1 were important to the extent that they influenced the cumulative dose.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Foot Diseases/chemically induced , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Pain/chemically induced , Peripheral Nervous System Diseases/chemically induced , Vincristine/adverse effects , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Cyclophosphamide/administration & dosage , Cytarabine/administration & dosage , Doxorubicin/administration & dosage , Etoposide/administration & dosage , Female , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Ifosfamide/administration & dosage , Infant , Male , Methotrexate/administration & dosage , Middle Aged , Multivariate Analysis , Vincristine/administration & dosage
14.
Invest New Drugs ; 14(1): 49-54, 1996.
Article in English | MEDLINE | ID: mdl-8880393

ABSTRACT

The taxanes are a new group of anticancer agents with a novel mechanism of action. They promote microtubule assembly and stabilize the microtubules. Paclitaxel (Taxol), the first agent in this group in clinical trials was isolated from the Pacific yew, Taxus brevifolia in 1971. Both in preclinical and clinical studies, paclitaxel and its semisynthetic analog docetaxel exhibit significant antitumor activity. This review will provide an overview of the clinical experience with the group of anti-microtubular agents, the taxanes in pediatric oncology.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Bridged-Ring Compounds/therapeutic use , Microtubules/drug effects , Neoplasms/drug therapy , Taxoids , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Bridged-Ring Compounds/pharmacokinetics , Bridged-Ring Compounds/pharmacology , Child , Clinical Trials, Phase I as Topic , Humans , Neoplasms/pathology , Neoplasms/ultrastructure
15.
Pediatr AIDS HIV Infect ; 6(5): 292-4, 1995 Oct.
Article in English | MEDLINE | ID: mdl-11361394

ABSTRACT

Malignancy occurs in 2-4% of children infected with human immunodeficiency virus (HIV). The most commonly reported type is non-Hodgkin's lymphoma. We describe the first reported case of chronic myelogenous leukemia (CML) in a child infected with HIV. The initial diagnosis was consistent with Philadelphia chromosome positive acute lymphocyte leukemia. However, on review of the patient's history and the persistence of the Philadelphia chromosome, the diagnosis of CML was made. CML needs to be added to the list of malignancies seen in patients with HIV.


Subject(s)
HIV Infections/complications , Infectious Disease Transmission, Vertical , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/complications , Child , Diagnostic Errors , Fatal Outcome , Female , HIV Infections/transmission , Humans , Immunophenotyping , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis
16.
J Immunother Emphasis Tumor Immunol ; 16(2): 125-31, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7804527

ABSTRACT

In preclinical studies, synergy was observed between tumor necrosis factor-alpha (TNF-alpha) and agents that interact with DNA topoisomerase II, such as actinomycin D (Act D). Based upon this, a Phase I study was conducted in pediatric patients utilizing an escalating dose of recombinant TNF (rTNF) in combination with a fixed dose of Act D. Act D (15 micrograms/kg/day) was administered daily by intravenous push immediately followed by intravenous rTNF daily for 5 consecutive days. Thirty-three patients with refractory malignancies were entered in the study, of whom 28 patients could be evaluated for toxicity. Malignancies included sarcomas (16), Wilms' tumor (6), leukemias (3), and others (3). The starting dose for rTNF was 40 micrograms/m2/day x 5 and was escalated in subsequent patient groups until nonhematopoietic, dose-limiting toxicity occurred. At 240 micrograms/m2/day of rTNF, three of six patients experienced grade 4 toxicity consisting of hypotension, hemorrhagic gastritis, and renal and liver biochemical abnormalities. Evidence of antitumor response was observed in two patients: one with metastatic Ewing's sarcoma and one with Wilms' tumor. We conclude that the maximum tolerated dose of rTNF when combined with Act D is between 200 and 220 micrograms/m2/day x 5 for pediatric patients.


Subject(s)
Dactinomycin/therapeutic use , Neoplasms/therapy , Tumor Necrosis Factor-alpha/therapeutic use , Adolescent , Adult , Child , Child, Preschool , Combined Modality Therapy , Dactinomycin/adverse effects , Female , Humans , Male , Tumor Necrosis Factor-alpha/adverse effects
17.
Echocardiography ; 11(2): 111-7, 1994 Mar.
Article in English | MEDLINE | ID: mdl-10150563

ABSTRACT

Echocardiography now permits tracking of the blood-endocardial border and automatic measurement of ventricular area throughout the cardiac cycle. To determine the accuracy of this technique, we compared echocardiographic measurements of area with similar measurements made by radionuclide technique in 19 children, ages 4-24 years (mean 13 years). The blood-endocardial border was tracked from the apical two-chamber view and radionuclide measurements were made from the left anterior oblique view. We measured echocardiographic end-diastolic area, end-systolic area, and fractional area change from the average of five cardiac cycles. The radionuclide area measurements were made from a gated blood pool study incorporating 700-1200 cardiac cycles. Results were compared by bias analysis. The mean differences (+/- 1 S.D.) between left ventricular area measurements were: end-diastole 1.13 +/- 2.3 cm2, end-systole -0.90 +/- 1.33 cm2, and fractional area change 7.4 +/- 9.3 (%). Differences between the measurements were within the limit of agreement (mean +/- 2 S.D.) in 55 of 57 measurements. The area measurements were not free from bias; the mean differences of area measurements were significantly different from zero for end-diastolic area (P < or = 0.05), end-systolic area (P < or = 0.01), and fractional area change (P < or = 0.002). Echocardiography tended to underestimate end-diastolic area and fractional area change and it tended to overestimate end-systolic area. Real-time tracking of the blood-endocardial border is possible and allows accurate measurement of ventricular area.


Subject(s)
Echocardiography/methods , Gated Blood-Pool Imaging/methods , Heart/diagnostic imaging , Image Processing, Computer-Assisted , Signal Processing, Computer-Assisted , Ventricular Function, Left , Adolescent , Adult , Antibiotics, Antineoplastic/toxicity , Child , Coronary Circulation/physiology , Echocardiography/instrumentation , Gated Blood-Pool Imaging/instrumentation , Humans , Myocardial Contraction/physiology , Neoplasms/drug therapy , Sensitivity and Specificity , Ventricular Function/physiology , Ventricular Function, Left/physiology
19.
J Clin Oncol ; 10(8): 1310-6, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1634921

ABSTRACT

PURPOSE: A phase II trial that uses liposome-encapsulated muramyl tripeptide phosphatidylethanolamine (L-MTP-PE) in patients with relapsed osteosarcoma is underway. To determine if in vivo cytokine induction plays a role in the mechanism of action of L-MTP-PE, we investigated the circulating cytokine levels of 16 patients who were undergoing therapy. PATIENTS AND METHODS: Patients had histologically proven osteosarcoma and pulmonary metastases that developed either during adjuvant chemotherapy or that were present at diagnosis and persisted despite chemotherapy. Patients were rendered disease-free by surgery. The major goal of the study was to improve the disease-free interval in this high-risk group. L-MTP-PE 2 mg/m2 was infused during a 1-hour period twice a week for 12 weeks, then once a week for 12 weeks. Serial blood samples were collected after L-MTP-PE administration and were assayed for cytokine levels (tumor necrosis factor-alpha [TNF alpha] interleukin-1 alpha [IL-1 alpha], IL-1 beta, IL-6, interferon-gamma [IFN-gamma], neopterin, C-reactive protein). RESULTS: After the infusion of L-MTP-PE, there was rapid induction of circulating TNF alpha and IL-6. TNF alpha levels peaked 1 to 2 hours after infusion in 10 of 16 patients, whereas peak IL-6 levels were detected at 2 to 3 hours in all patients. Induction of circulating TNF alpha and IL-6 was evident only after the first dose of L-MTP-PE. Neither IL-1 alpha nor IL-1 beta was detected in the plasma. Neopterin levels increased at 24 hours postinfusion, which indicated macrophage activation, and were not related to the induction of circulating IFN-gamma. C-reactive protein was elevated in all patients at 24 hours and decreased by 72 hours. Unlike circulating TNF alpha and IL-6, elevations in C-reactive protein and neopterin could be detected throughout the treatment course. CONCLUSION: It is concluded that L-MTP-PE has specific biologic effects in patients with osteosarcoma that may be important to the drug's immunostimulatory capacity and its effectiveness as an antitumor agent.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/analogs & derivatives , Antineoplastic Agents/pharmacology , Cytokines/drug effects , Monocytes, Activated Killer/drug effects , Osteosarcoma/drug therapy , Osteosarcoma/immunology , Phosphatidylethanolamines/pharmacology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Acetylmuramyl-Alanyl-Isoglutamine/therapeutic use , Antineoplastic Agents/therapeutic use , Biopterins/analogs & derivatives , Biopterins/blood , C-Reactive Protein/drug effects , Cytokines/blood , Drug Carriers , Drug Evaluation , Humans , Liposomes , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Neopterin , Osteosarcoma/secondary , Phosphatidylethanolamines/therapeutic use
20.
J Pediatr ; 120(6): 987-93, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1593362

ABSTRACT

To evaluate the safety, tolerance, and pharmacokinetics of fluconazole in children with neoplastic diseases, we studied fluconazole in 26 children, aged 5 to 15 years, with normal renal function who were receiving treatment for cancer. The patients received fluconazole, 2, 4, or 8 mg/kg per day for 7 days intravenously for a 2-hour period. Patients had no nausea or vomiting related to fluconazole; three patients had an asymptomatic rise in hepatic aminotransferase values after four to six doses (one patient at 2 mg/kg per day and two patients at 8 mg/kg per day), which returned to normal within 2 weeks after discontinuation of the drug. Fluconazole showed linear first-order kinetics over the dosage range tested and during multiple dosing. After the first dose, mean clearance was 22.8 +/- 2.3 ml/min, volume of distribution 0.87 +/- 0.06 L/kg, and terminal elimination half-life 16.8 +/- 1.1 hours. Similarly, after the last dose, clearance was 19.4 +/- 1.3 ml/min, volume of distribution 0.84 +/- 0.04 L/kg, and terminal elimination half-life 18.1 +/- 1.2 hours. Patients receiving their first fluconazole dose of 8 mg/kg achieved peak serum levels of 9.5 +/- 0.4 microgram/ml and trough levels of 2.7 +/- 0.5 microgram/ml 24 hours later, and an area under the serum concentration-time curve from time zero to infinity of 186 +/- 16 micrograms.hr per milliliter. Renal clearance of fluconazole was 65% +/- 5% of total clearance and demonstrated the predominantly renal excretion of this drug. We suggest that the shorter serum half-life and the higher frequency of aminotransferase elevations in comparison with those of adults warrant careful investigation of fluconazole in controlled clinical trials.


Subject(s)
Fluconazole/pharmacokinetics , Fluconazole/toxicity , Leukemia, Myeloid, Acute/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Soft Tissue Neoplasms/metabolism , Child , Drug Administration Schedule , Drug Evaluation , Female , Humans , Kidney/metabolism , Liver/metabolism , Liver Function Tests , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...