Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Med Pediatr Oncol ; 37(4): 390-2, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11568904

ABSTRACT

BACKGROUND: The prognosis of children with high-grade astrocytomas of the central nervous system is grim and has not been substantially improved by conventional chemoradiotherapy. We performed a multi-institutional phase I study to determine the toxicities and tolerance of concurrent external beam radiation of the brain and a unique dose-schedule of paclitaxel as a radiation sensitizer. PROCEDURE: Paclitaxel was delivered intravenously as a continuous 24 h/day, 7 days/week infusion during the entire 6-week course of fixed schedule standard radiation therapy. The dose of paclitaxel was escalated in patient cohorts in standard phase I design. RESULTS: Eleven patients (eight brain stem gliomas, one glioblastoma multiforme, and two gliomatosis cerebri) were treated. Dose-limiting toxicity was encountered in the two patients treated at 6 mg/(m(2)/24 h), both of whom developed severe obstipation requiring prolonged hospitalization. CONCLUSIONS: We have shown in this first study of its kind that paclitaxel can be administered safely to children as a 6-week continuous infusion concurrent with cranial irradiation. The maximally tolerated and recommended phase II dose is 4 mg/(m(2)/day). The benefits of taxanes as clinical radiation sensitizers for children with high-grade gliomas, if any, remain to be determined.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Paclitaxel/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Radiotherapy, High-Energy/methods , Adolescent , Brain/radiation effects , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Child , Child, Preschool , Combined Modality Therapy , Drug Administration Schedule , Female , Follow-Up Studies , Glioblastoma/drug therapy , Glioblastoma/mortality , Glioblastoma/pathology , Glioblastoma/radiotherapy , Glioma/drug therapy , Glioma/mortality , Glioma/pathology , Glioma/radiotherapy , Humans , Infusions, Intravenous , Male , Radiation Injuries/prevention & control , Survival Analysis , Treatment Outcome
2.
J Pediatr Hematol Oncol ; 23(5): 277-81, 2001.
Article in English | MEDLINE | ID: mdl-11464982

ABSTRACT

PURPOSE: To assess the efficacy and define the toxicity of paclitaxel given at a dosage of 350 mg/m2 every 3 weeks as a 24-hour continuous infusion to children with recurrent or progressive primary brain tumors. PATIENTS AND METHODS: Seventy-three eligible patients, ages 4 months to 19 years, with progressive or recurrent primary brain tumors were treated according to a Pediatric Oncology Group (POG) phase II protocol with paclitaxel (POG 9330). Tumor histologic strata included: astrocytoma (n = 4), malignant glioma (n = 13), medulloblastoma (n = 16), brain stem glioma (n = 15), ependymoma (n = 13), and miscellaneous histologies (n = 12). All patients had previous histologic confirmation of a primary intracranial or spinal cord tumor with magnetic resonance imaging or computed tomography documentation of unequivocally measurable progressive or recurrent disease. All patients had received previous therapy including surgery, radiation therapy, and/or chemotherapy, but no patient had been previously treated on more than one phase II trial. Paclitaxel was administered as a 24-hour intravenous infusion at a dosage of 350 mg/m2 every 3 weeks. Neurologic and neuroradiologic reevaluations were performed after every second course. Patients were allowed to continue therapy for a total of 18 cycles in the absence of progressive disease or unacceptable toxicity. RESULTS: Seventy-five patients were enrolled onto the POG 9330 protocol; two ineligible patients were removed from the study before receiving any therapy. Of the 73 eligible patients, 72 were evaluable for toxicity and 70 were either fully or partially evaluable for disease response. There was one complete response and three partial responses (5.7%). Twenty patients had stable disease for more than 2 months. Toxicities included mild nausea, central nervous system toxicity, myelosuppression, and febrile neutropenia, including one septic death. One grade 2 and two grade 3 allergic reactions occurred. No cardiac toxicities or arthralgias were reported. CONCLUSION: Paclitaxel is well tolerated in children with recurrent or progressive brain tumors at this dosage and schedule and may result in short-term disease stabilization in this patient population. The lack of a significant number of patients with measurable disease regression, however, precludes it from being identified as an active agent when administered as a single agent by 24-hour continuous infusion.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Brain Neoplasms/drug therapy , Paclitaxel/therapeutic use , Adolescent , Antineoplastic Agents, Phytogenic/adverse effects , Astrocytoma/drug therapy , Astrocytoma/pathology , Child , Child, Preschool , Dexamethasone/therapeutic use , Disease Progression , Drug Hypersensitivity/prevention & control , Ependymoma/drug therapy , Ependymoma/pathology , Female , Glioma/drug therapy , Glioma/pathology , Humans , Immunosuppressive Agents/therapeutic use , Infant , Infratentorial Neoplasms/drug therapy , Infratentorial Neoplasms/pathology , Infusions, Intravenous , Male , Medulloblastoma/drug therapy , Medulloblastoma/pathology , Nausea/chemically induced , Neoplasm Recurrence, Local , Neutropenia/chemically induced , Paclitaxel/adverse effects , Remission Induction , Salvage Therapy , Treatment Failure
5.
Ophthalmology ; 103(12): 1989-97, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9003332

ABSTRACT

PURPOSE: Management of intraocular retinoblastoma was initiated with 2-drug chemotherapy in an effort to improve the rate of vision preservation and ocular salvage and to avoid or delay the use of external beam radiation treatment. METHODS: Six patients with intraocular retinoblastoma (five bilateral; one unilateral, 1 month old) received 6 to 7 monthly cycles of intravenously administered carboplatin and etoposide (VP-16) as primary treatment. No eyes were enucleated primarily. Twelve of the 33 discrete tumors in the 11 study eyes received prophylactic supplemental treatment with cryotherapy or laser hyperthermia. Response was documented with frequent eye examinations with the patient under general anesthesia and with repeated fundus photography. RESULTS: All eight larger tumors (> 10-mm diameter) underwent dramatic regression after treatment with chemotherapy alone, and six of these tumors ultimately became fully calcific. One larger tumor and two smaller tumors showed post-treatment growth, each within 2 months after completion of chemotherapy. Six larger tumors were observed without growth or further treatment for 7 to 21 months after completion of chemotherapy. Subretinal fluid resorbed completely in four of four eyes with extensive retinal detachment, and vitreous seeding diminished considerably in four of four eyes. In five eyes, intraocular disease recurrence at a distance from any initially observed tumor eventually required treatment with external beam radiation (three eyes) or enucleation (three eyes). Eight of 11 involved eyes were salvaged, including 5 of 8 with larger tumors and 4 of 4 with vitreous seeding; 4 retained eyes received no radiation exposure, including 3 with larger tumors and 1 with vitreous seeding. Good vision was preserved in six eyes, two of which were markedly improved after occlusion therapy for amblyopia. There was no extraocular disease recurrence and no serious harm from treatment during observation ranging from 12 to 40 months after diagnosis. CONCLUSION: Chemotherapy with carboplatin and etoposide shows promise as initial treatment for intraocular retinoblastoma. Further study is indicated to define its proper role in the management of this disease.


Subject(s)
Antineoplastic Agents/therapeutic use , Carboplatin/therapeutic use , Etoposide/therapeutic use , Eye Neoplasms/drug therapy , Retinoblastoma/drug therapy , Antineoplastic Agents/administration & dosage , Carboplatin/administration & dosage , Child, Preschool , Cryotherapy , Etoposide/administration & dosage , Eye Enucleation , Eye Neoplasms/pathology , Eye Neoplasms/physiopathology , Female , Fundus Oculi , Humans , Hypothermia, Induced , Infant , Infusions, Intravenous , Male , Neoplasm Recurrence, Local , Radiotherapy, Adjuvant , Remission Induction , Retinoblastoma/pathology , Retinoblastoma/physiopathology
6.
Lab Invest ; 72(4): 419-23, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7723280

ABSTRACT

BACKGROUND: Central nervous system teratomas are a rare subgroup of extragonadal germ cell tumors. Previous studies show ovarian mature teratomas to be karyotypically normal; those with increasing immaturity show increasing cytogenetic abnormalities. Adult testicular teratomas, regardless of maturity, most often show the i sochromosome 12p, a consistent chromosomal abnormality seen in adult testicular germ cell tumors. This study investigates the cytogenetic abnormalities of six central nervous system teratomas by the use of routine karyotypic analysis and fluorescence in situ hybridization. EXPERIMENTAL DESIGN: Karyotypic analysis was performed on two tumors. Four additional tumors were analyzed by dual-labeled fluorescence in situ hybridization. Paraffin blocks were disaggregated, and nuclei were hybridized with both biotin- and digoxigenin-labeled probes to the centromeric domains of the X and Y chromosomes, respectively. Tumor cells were scored for X and Y copy number. Tumor ploidy was determined by image analysis of the disaggregated specimens. RESULTS: The patients (five male, one female) ranged from 2 to 25 years of age. All presented with a solitary mass in the suprasellar region. A mature teratoma was 48, XXYY; an immature teratoma was 47, XXY, dir dup 11 (q12-22). Lymphocyte analysis of the second patient showed a normal constitutional karyotype. Fluorescence in situ hybridization analysis of four additional patients showed both one mature and one immature teratoma from male hosts to have 2X and 1Y signals in the majority of the cells. An immature teratoma from a male host showed 2X and 2Y signals. The above three tumors were diploid by static image analysis. An immature teratoma from a female host showed multiple (four to six) X signals in more than 70% of the cells. Ploidy analysis was unavailable for this case. CONCLUSIONS: Although patients with Klinefelter syndrome (47,XXY) and patients with 46,XY gonadal dysgenesis show an increased incidence of germ cell tumors, numerical sex chromosomal abnormalities have not been described in extragonadal teratomas. Our results support a role for sex chromosomes in the development of central nervous system teratomas.


Subject(s)
Brain Neoplasms/genetics , Chromosome Aberrations , Chromosome Disorders , Pineal Gland , Sex Chromosomes , Teratoma/genetics , Adult , Child , Child, Preschool , Cytogenetics/methods , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Ploidies
7.
J Clin Oncol ; 10(12): 1857-64, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1453200

ABSTRACT

PURPOSE: The purpose of this study was to determine the toxicities of and responses to high-dose busulfan and cyclophosphamide with autologous bone marrow transplant (ABMT) in patients with recurrent or refractory pediatric solid tumors. PATIENTS AND METHODS: We treated 18 patients (ages, 2 to 38 years; median, 14) who had tumors that were resistant to conventional chemotherapy and radiotherapy with busulfan 16 mg/kg and cyclophosphamide 200 mg/kg. Seventeen patients received bone marrow purged with 4-hydroperoxycyclophosphamide; one received unpurged marrow. RESULTS: Despite extensive prior treatment, including radiotherapy in 16 patients, toxicity generally was acceptable. For seven patients with measurable disease, there were three partial responses of 2, 10, and 20 months' duration, three patients with stable disease (SD), and one early, toxic death. Of the 11 patients with no measurable disease at the time of transplantation, one patient with osteosarcoma continues in remission at 57+ months and one third of the patients survived for at least 16 months. Mucositis was the predominant nonhematopoietic toxicity. CONCLUSION: Although the high-dose busulfan and cyclophosphamide combination showed modest activity, changes in the preparative regimen should be considered to improve the response rate in refractory tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Transplantation , Neoplasms/therapy , Adolescent , Adult , Bone Marrow Purging , Busulfan/administration & dosage , Child , Child, Preschool , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Humans , Recurrence , Survival Analysis , Transplantation, Autologous
8.
Pediatrics ; 89(5 Pt 1): 895-7, 1992 May.
Article in English | MEDLINE | ID: mdl-1579400

ABSTRACT

It is often difficult to measure cerebrospinal fluid (CSF) pressure in children. CSF flow through a spinal needle is described by the equation: Flow = pressure/(needle constant x relative viscosity). Thus, CSF flow rate during lumbar puncture can be used to estimate CSF pressure. Because the viscosity of CSF is approximately the same as that of normal saline, 0.9% NaCl was used to model CSF flow in vitro. Flow of saline through various spinal needles was measured as pressure and temperature were varied to determine needle constants and variation in viscosity with temperature. Counting periods for which the number of drops counted equals the pressure (in centimeters of H2O) then were determined for each needle size. At patient temperatures less than 40 degrees C, counting periods were calculated at 21, 39, and 12 seconds, for 22-gauge 1.5-inch, 22-gauge 3.5-inch, and 20-gauge 3.5-inch spinal needles, respectively. Viscosity decreased slightly above 40 degrees C, and counting periods became 20, 37, and 11 seconds. Finally, the method was tested prospectively in 12 patients by comparing drop count (over the calculated counting period) to manometric pressure measurement. Drop counts were within 15% of manometric pressure in all patients. This method allows simple and rapid estimation of CSF pressure during lumbar puncture.


Subject(s)
Cerebrospinal Fluid Pressure/physiology , Needles , Spinal Puncture , Child , Evaluation Studies as Topic , Humans , Manometry , Prospective Studies , Rheology , Time Factors , Viscosity
9.
Exp Hematol ; 19(8): 734-41, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1714400

ABSTRACT

Initiation of DNA synthesis by recombinant colony-stimulating factors (CSFs) was assessed in normal human marrow blast cells isolated by expression of CD34 antigen (tritiated thymidine incorporation). Continuous exposure to CSF was required. A mild increase in DNA synthesis was initiated by granulocyte CSF (G-CSF; greater than or equal to 1 ng/ml), to approximately 1.5 times control levels. A greater increase was initiated by granulocyte-macrophage CSF (GM-CSF), with a threshold of approximately 0.1 ng/ml and a plateau increment 2.5 times control levels. CD34+ cells were stimulated by interleukin 3 (IL-3) over a wide concentration range: two times control at 0.1/ml, three times control at 1 ng/ml, and four times control at 10 ng/ml. Overlap between responding populations was analyzed. G-CSF plus GM-CSF induced DNA synthesis greater than GM-CSF alone and supported the growth of much larger granulocyte-monocyte colonies. At saturating IL-3 concentrations, neither G-CSF nor GM-CSF induced additional DNA synthesis; at lower concentrations of IL-3, however, GM-CSF recruited additional cells into DNA synthesis. Using CD10 and CD19 antibodies to separate B-lineage cells, the CD34+ cells responding to CSF were observed to be in the non-B-lineage subset. Therefore 1) the response of CD34+ cell subsets CSFs is IL-3 greater than GM-CSF greater than G-CSF, and the IL-3-responsive population is heterogeneous for dose requirement; 2) a CD34+ subpopulation responding to concurrent G-CSF and GM-CSF includes increased proliferative potential cells; 3) IL-3-responsive cells include GM-CSF- and G-CSF-responsive cells, but cells responding to lower IL-3 concentration do not respond to GM-CSF; and 4) B-cell precursors do not respond to GM-CSF or IL-3 in this assay.


Subject(s)
Antigens, CD/analysis , Bone Marrow Cells , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Interleukin-3/pharmacology , Antigens, CD19 , Antigens, CD34 , Antigens, Differentiation/analysis , Antigens, Differentiation, B-Lymphocyte/analysis , Antigens, Neoplasm/analysis , Cell Division/drug effects , DNA/biosynthesis , Dose-Response Relationship, Drug , Drug Synergism , Humans , In Vitro Techniques , Neprilysin , Recombinant Proteins
10.
J Neurooncol ; 11(1): 57-63, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1919647

ABSTRACT

Sixteen patients with recurrent childhood brain tumors were treated with intravenous cisplatin, cytosine arabinoside and etoposide (PAE), daily for three days every three to four weeks. Objective responses were observed in 6 of 15 evaluable patients and an additional six patients had stable disease for greater than 6 months. The tumor-specific response rate for astrocytoma/glioma was 3 of 7 and for medulloblastoma was 2 of 4. The mean progression-free interval was 11.0 months and the hazard rate for progression was 0.085 per patient-month of observation. The most common toxicities were neutropenia and thrombocytopenia. Clinically significant ototoxicity was identified in 7 patients. The activity of PAE chemotherapy for recurrent childhood brain tumors warrants further investigation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/toxicity , Brain Neoplasms/drug therapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Astrocytoma/drug therapy , Cerebellar Neoplasms/drug therapy , Child , Cisplatin/administration & dosage , Cytarabine/administration & dosage , Drug Evaluation , Ependymoma/drug therapy , Etoposide/administration & dosage , Female , Glioma/drug therapy , Humans , Male , Medulloblastoma/drug therapy , Neoplasm Recurrence, Local , Pinealoma/drug therapy , Probability , Prognosis
11.
Med Pediatr Oncol ; 19(1): 16-21, 1991.
Article in English | MEDLINE | ID: mdl-1990253

ABSTRACT

Neuraxis radiation therapy (RT) for primary intracranial tumors is associated with major late effects if administered to very young children. To control residual tumor and to delay RT, we treated eight young children (median age 6.5 months) with primary central nervous system (CNS) tumors using combination chemotherapy: cisplatin, 20 mg/M2/day plus VP-16, 75 mg/M2/day i.v. for 5 days, given q. 3-6 weeks for 8 cycles. The tumors were medulloblastoma (one), malignant ependymoma (two), primitive neuroectodermal tumor PNET (two), malignant glioma (two), astrocytoma (one). Six had measurable disease; three had positive cerebrospinal fluid (CSF) cytopathology. All patients with measurable tumor had initial objective responses (three) complete response [CR], one partial response [PR], two minor response [MR], including cytopathology (three CR of three) and metastatic deposits (two CR of two). One patient relapsed during chemotherapy. Median time to disease progression was 17.5 months; median survival was 34 months. Three patients, none of whom received RT, have prolonged progression-free intervals of 47-67 months to date. Neurodevelopmental progress continued during and after chemotherapy. Chemotherapy toxicity was mild. Median neutrophil nadir was 312/mm3, platelets 72,000. Fever during neutropenia occurred in six of 61 courses. Moderate high-frequency auditory losses were detected in three patients, and mild renal injury (GFR less than 70 ml/min) was detected in two of seven evaluable children. This pilot study demonstrates the apparent efficacy and mild toxicity of 5 day courses of cisplatin plus VP-16, with delayed RT, in young children with CNS neoplasms. A POG treatment protocol that incorporates cisplatin plus VP-16 is evaluating primary chemotherapy with delayed radiotherapy in larger numbers of pediatric brain tumor patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Cisplatin/administration & dosage , Etoposide/administration & dosage , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/surgery , Child, Preschool , Cisplatin/adverse effects , Combined Modality Therapy , Etoposide/adverse effects , Female , Humans , Infant , Male , Pilot Projects , Tomography, X-Ray Computed
12.
Am J Pediatr Hematol Oncol ; 13(2): 217-21, 1991.
Article in English | MEDLINE | ID: mdl-1712550

ABSTRACT

Complete yet nontoxic removal of tumor cells from autologous marrow grafts has proved difficult. New methods for separating normal stem cells from tumor cells are needed. The CD34+ cells in bone marrow, 1-2% of the low-density leukocytes, include precursors of all lymphohematopoietic lineages and probably also the primitive cells responsible for engraftment. A nontoxic, inexpensive, reproducible, and clinically applicable method for positive selection of CD34+ cells was developed. Paramagnetic microspheres coated with goat anti-mouse IgG1 are used to partition the cells; brief incubation with chymopapain is used to release them from the beads. Chymopapain exposure does not injury colony-forming cells or delay engraftment in rodents. Clinical volumes of bone marrow can be processed rapidly. In pilot experiments, the resulting grafts have a purity of 85-99% CD34+ cells and 40% median recovery of the assayable colony-forming cells. These studies form the background for a Phase I trial of autologous BMT using CD34+ stem cells.


Subject(s)
Antigens, CD/immunology , Bone Marrow Transplantation/immunology , Cell Separation/methods , Hematopoietic Stem Cells/immunology , Animals , Antigens, CD34 , Hematopoietic Stem Cell Transplantation , Humans , Magnetics , Microspheres , Rats
13.
Hematol Oncol Clin North Am ; 4(4): 849-65, 1990 Aug.
Article in English | MEDLINE | ID: mdl-2228899

ABSTRACT

The interrelationship between proliferation, differentiation, and activation responses of hematopoietic progenitor cells and mature blood cells is complex. Therefore, we are only now learning what role colony-stimulating factors play in the regulation of normal hematopoiesis in vivo and in the dysregulation of hematopoiesis in leukemia. Recent advantages in molecular hematology have opened the door to the therapeutic administration of recombinant growth factors. Through continued preclinical trials in animals and by clinical trials in humans, a better understanding of the precise target cells and mechanisms of action of hematopoietic growth factors will improve the therapeutic index of administering colony-stimulating factors. Better understanding of hematopoietic growth factors will, in turn, suggest novel approaches to therapy of acute lymphoblastic leukemia.


Subject(s)
Growth Substances/physiology , Hematopoiesis/immunology , Leukemia/etiology , Lymphocytes/immunology , Colony-Stimulating Factors/therapeutic use , Growth Substances/therapeutic use , Humans , Leukemia/drug therapy , Leukemia/immunology
14.
Pediatr Neurol ; 6(3): 163-70, 1990.
Article in English | MEDLINE | ID: mdl-2193641

ABSTRACT

Thirteen children (1.8-15.8 years of age) with brain tumors were studied with [11C]L-methionine positron emission tomography (METPET). Patients were injected intravenously with tracer before a baseline PET scan was obtained. To assess the sensitivity of [11C]L-methionine uptake to competitive inhibition, 10 patients received oral L-phenylalanine (100 mg/kg); 1 hour later, a second METPET was obtained. Subjective assessment of [11C]L-methionine uptake closely paralleled results of quantitative examination (r = 0.81). [11C]L-methionine uptake in tumor-containing brain was increased in 11 patients (mean ratio of [11C] radioactivity in tumor to normal brain: 1.5 +/- 0.57; range: 1.13-2.98). Increased tracer uptake occurred in ependymomas (3), medulloblastoma (1), and astrocytomas (5), but was less intense in low-grade tumors. L-phenylalanine reduced L-methionine uptake (25-69%) in 70% of studies. L-methionine uptake was not sensitive to competitive inhibition in brain radiation injury. Two-phase METPET is of potential value in difficult clinical situations evident in children with brain tumors, including the differential diagnosis of tumor recurrence and cerebral radiation injury.


Subject(s)
Astrocytoma/diagnostic imaging , Brain Neoplasms/diagnostic imaging , Ependymoma/diagnostic imaging , Medulloblastoma/diagnostic imaging , Tomography, Emission-Computed/methods , Adolescent , Child , Child, Preschool , Diagnosis, Differential , Female , Humans , Infant , Male , Methionine , Neoplasm Recurrence, Local/diagnostic imaging , Phenylalanine , Radiation Injuries/diagnostic imaging , Sensitivity and Specificity
15.
J Clin Oncol ; 8(2): 280-6, 1990 Feb.
Article in English | MEDLINE | ID: mdl-2299371

ABSTRACT

We evaluated the courses of 115 consecutive cases of pediatric acute leukemia treated with induction chemotherapy. Seventy-two patients developed fever associated with neutropenia; 15 developed systemic fungal infections. We reviewed multiple demographic and treatment characteristics of these patients in an attempt to identify potential risk factors for the development of invasive fungal disease (IFD). Risk factors identified in a univariate analysis included duration of neutropenia after first fever (P less than .0001), diagnosis of acute nonlymphocytic leukemia (ANLL) (P = .003), onset of fever and neutropenia within 5 days of starting induction chemotherapy (P = .009), and multiple (greater than one) surveillance culture sites positive for fungal organisms (P = .02). In a multiple logistic regression analysis, duration of neutropenia (P less than .001) remained a significant risk factor. The study group of patients had a significantly higher risk of fungal infections than a matched group of leukemia patients developing fever with neutropenia due to postremission consolidation chemotherapy (P = .003). In the first 48 patients, 14 (29%) developed IFD. In the subsequent patients (n = 24), intravenous miconazole (5 mg/kg every 8 hours) was begun at the time of the first fever. One of the 24 patients (4%) given miconazole developed IFD. The use of miconazole was a negative risk factor for the development of IFD in univariate (P = .01) and multivariate (P = .05) analysis. We conclude that pediatric leukemia patients who develop fever associated with neutropenia during induction chemotherapy are at high risk for developing IFD. The role of intravenous miconazole at the time of the first fever in this group deserves further study.


Subject(s)
Agranulocytosis/complications , Fever/complications , Leukemia, Myeloid, Acute/drug therapy , Mycoses/etiology , Neutropenia/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Humans , Infant , Leukemia, Myeloid, Acute/complications , Miconazole/therapeutic use , Multivariate Analysis , Mycoses/prevention & control , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Regression Analysis , Remission Induction , Risk Factors
16.
Prog Clin Biol Res ; 333: 387-401; discussion 402, 1990.
Article in English | MEDLINE | ID: mdl-1689854

ABSTRACT

Immunologic strategies for removal of malignant cells from autologous marrow grafts by "negative selection" (i.e., "purging") requiring multiple specific monoclonal antibodies for each tumor type. "Positive selection" of marrow stem cells for grafting is a possible alternative strategy, using a monoclonal antibody which selectively recognizes lymphohematopoietic stem cells. The human hematopoietic progenitor cell antigen, CD34, is an integral cell membrane glycoprotein of approximately 115 kD, which has been molecularly cloned and sequenced. Although its function has not been determined, the glycoprotein has been characterized biochemically, including preliminary epitope mapping. Collective results from several laboratories indicate that CD34 monoclonal antibodies (My10, BI-3C5, 12.8, etc.) have the appropriate specificity to warrant testing their utility in positive selection for autologous bone marrow transplantation. First, precursors for all human hematopoietic lineages assayed (including most CFU-GM, BFU-E, CFU-MEG, CFU-EO, CFU-MIX or CFU-GEMM, pre-CFU, CFUBLAST, and terminal transferase+ B [and probably T] lymphoid precursors) are CD34+. Second, only 1.5% (mean) of low density human marrow mononuclear cells express CD34; mature human blood and marrow cells are CD34-. Endothelial cells are the only fixed tissue cells which express CD34. Third, the expression of CD34 in malignancies appears to parallel normal cellular expression: of hematopoietic malignancies, some acute leukemias and chronic myelogenous leukemia blasts are CD34+, but chronic lymphois leukemias, lymphomas, myelomas and non-hematopoietic malignancies are uniformly CD34-. Fourth, it appears feasible to isolate CD34+ cells from clinical marrow harvest samples in large scale, using either columns or immunomagnetic microspheres. Fifth, recent studies in very small numbers of non-human primates and human patients suggest that isolated CD34+ cells include the true hematopoietic stem cell, since transplantation of CD34+ cells, into myeloblated recipients results in at least short-term hematopoietic engraftment. It is anticipated that transplantation of CD34+ marrow cells may have broad applicability in clinical bone marrow transplantation.


Subject(s)
Bone Marrow Transplantation/methods , Cell Separation/methods , Hematopoietic Stem Cells , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Antigens, CD34 , Antigens, Differentiation/immunology , Antigens, Surface/immunology , Cell Fractionation , Chymopapain , Colony-Forming Units Assay , Hematopoietic Stem Cells/immunology , Humans , Light , Magnetics , Microspheres , Monocytes/immunology , Rats , Scattering, Radiation
18.
Am J Pediatr Hematol Oncol ; 11(3): 249-67, 1989.
Article in English | MEDLINE | ID: mdl-2675658

ABSTRACT

Understanding of the roles and molecular mechanisms of hematopoietic growth factors has increased greatly in recent years. This past decade has also brought us tantalizingly close to linking a group of genes normally involved in the regulation of growth and differentiation--the cellular proto-oncogenes--to the process of malignant transformation. In this article, we review the known actions of hematopoietic growth factors in normal hematopoiesis and in hematologic malignancies. We then discuss current concepts of the roles of proto-oncogenes in normal cells and their potential involvement in leukemogenic events. A merging of these concepts with classical hypotheses of multistage carcinogenesis is emphasized.


Subject(s)
Growth Substances/physiology , Hematopoietic Stem Cells , Leukemia/genetics , Proto-Oncogenes , Growth Substances/therapeutic use , Hematopoietic Cell Growth Factors , Humans , Leukemia/metabolism , Leukemia/therapy
19.
Am J Pediatr Hematol Oncol ; 11(2): 158-61, 1989.
Article in English | MEDLINE | ID: mdl-2546463

ABSTRACT

Leukemia accounts for 15-20% of the secondary malignancies among survivors of Wilms' tumor. We report three patients who developed leukemia after the successful treatment of Wilms' tumor, each of whom demonstrates the importance of close long-term medical surveillance. The first patient developed Philadelphia chromosome positive chronic myelogenous leukemia (CML) 6 years after the diagnosis of Wilms' tumor. This is the second report of CML occurring after Wilms' tumor. The other two patients developed acute nonlymphocytic leukemia (ANLL) 3 and 18 years after successful treatment of Wilms' tumor. In one patient, the clinical manifestations were subtle, and in the other the latency period was the longest reported for secondary leukemia following Wilms' tumor. We conclude that survivors of childhood cancer require frequent medical surveillance even in their adult years.


Subject(s)
Kidney Neoplasms , Kidney Neoplasms/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Acute/pathology , Neoplasms, Multiple Primary/pathology , Wilms Tumor/pathology , Alkylating Agents/adverse effects , Alkylating Agents/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Combined Modality Therapy , Humans , Kidney Neoplasms/therapy , Wilms Tumor/therapy
20.
J Clin Oncol ; 5(12): 1900-11, 1987 Dec.
Article in English | MEDLINE | ID: mdl-3500279

ABSTRACT

2'Deoxycoformycin (dCF) specifically inhibits adenosine deaminase (ADA) and causes selective cytotoxicity of normal and malignant T cells. In clinical trials, dCF caused rapid lysis of malignant T lymphoblasts. Although dCF has been associated with dose-limiting nonhematopoietic toxicities, myelosuppression has not been observed. Since dCF is relatively nontoxic to hematopoietic stem cells, we tested dCF for utility in the ex vivo purging of malignant T lymphoblasts from remission leukemic bone marrow for autologous bone marrow transplantation. We found that T lymphoblast cell lines were sensitive to dCF (plus deoxyadenosine [dAdo]) under conditions that did not ablate human hematopoietic colony-forming cells. Moreover, combined pharmacologic (dCF plus dAdo) and immunologic (anti-T cell monoclonal antibodies [McAb] plus complement) purging resulted in additive reduction in clonogenic T lymphoblasts. These results provide the basis for a clinical trial of bone marrow transplantation using combined pharmacologic/immunologic purging of T lymphoblasts from patients' harvested autologous marrow.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Agents/pharmacology , Coformycin/pharmacology , Colony-Forming Units Assay , Lymphoma/pathology , Ribonucleosides/pharmacology , T-Lymphocytes/drug effects , Cell Line , Coformycin/analogs & derivatives , Complement System Proteins/physiology , Deoxyadenosines/pharmacology , Humans , Lymphocyte Depletion , Pentostatin , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...