Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Blood ; 131(24): 2661-2669, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29724899

ABSTRACT

Pinometostat (EPZ-5676) is a first-in-class small-molecule inhibitor of the histone methyltransferase disrupter of telomeric silencing 1-like (DOT1L). In this phase 1 study, pinometostat was evaluated for safety and efficacy in adult patients with advanced acute leukemias, particularly those involving mixed lineage leukemia (MLL) gene rearrangements (MLL-r) resulting from 11q23 translocations. Fifty-one patients were enrolled into 6 dose-escalation cohorts (n = 26) and 2 expansion cohorts (n = 25) at pinometostat doses of 54 and 90 mg/m2 per day by continuous intravenous infusion in 28-day cycles. Because a maximum tolerated dose was not established in the dose-escalation phase, the expansion doses were selected based on safety and clinical response data combined with pharmacodynamic evidence of reduction in H3K79 methylation during dose escalation. Across all dose levels, plasma pinometostat concentrations increased in an approximately dose-proportional fashion, reaching an apparent steady-state by 4-8 hours after infusion, and rapidly decreased following treatment cessation. The most common adverse events, of any cause, were fatigue (39%), nausea (39%), constipation (35%), and febrile neutropenia (35%). Overall, 2 patients, both with t(11;19), experienced complete remission at 54 mg/m2 per day by continuous intravenous infusion, demonstrating proof of concept for delivering clinically meaningful responses through targeting DOT1L using the single agent pinometostat in MLL-r leukemia patients. Administration of pinometostat was generally safe, with the maximum tolerated dose not being reached, although efficacy as a single agent was modest. This study demonstrates the therapeutic potential for targeting DOT1L in MLL-r leukemia and lays the groundwork for future combination approaches in this patient population. This clinical trial is registered at www.clinicaltrials.gov as NCT01684150.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Histones/metabolism , Leukemia, Myeloid, Acute/drug therapy , Methyltransferases/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Benzimidazoles/adverse effects , Female , Histone-Lysine N-Methyltransferase , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Methylation/drug effects , Methyltransferases/metabolism , Middle Aged , Young Adult
2.
Pediatr Hematol Oncol ; 37(6): 465-474, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32338562

ABSTRACT

Background: Panobinostat demonstrates activity against pediatric cancers in vitro. A phase I trial in children with refractory hematologic malignancies was conducted. Study design: The trial evaluated two schedules of oral panobinostat using 3 + 3 dose escalations in 28-day cycles. For children with leukemia, panobinostat was given once daily three days a week each week at 24, 30 and 34 mg/m2/day. For children with lymphoma, panobinostat was given once daily three days a week every other week at 16, 20 and 24 mg/m2/day. Cerebrospinal fluid (CSF) from Day 29 of the first cycle, when available, was evaluated for PK. The study was registered on clinicaltrials.gov (NCT01321346) Results: Twenty-two subjects enrolled with leukemia. Five enrolled at dose level 1, 6 at dose level 2, and 11 at dose level 3. There was one dose limiting toxicity (DLT) in the leukemia arm at dose level 3 (Grade 4 hypertriglyceridemia), but no maximum tolerated dose (MTD) was identified. No subjects required removal from protocol therapy for QTc prolongation. PK studies were available in 11 subjects with similar exposure in children as in adults. Four Day 29 CSF specimens were found to have panobinostat levels below the lower limit of quantification. Five subjects with lymphoma were enrolled and received study drug, and 4 were evaluable for DLT. A DLT was reported (Grade 3 enteritis) on the lymphoma arm. Conclusions: Panobinostat was tolerated in heavily pretreated pediatric subjects. Gastrointestinal effects were observed on this study. There were no cardiac findings. There were no responses.


Subject(s)
Hematologic Neoplasms/drug therapy , Leukemia/drug therapy , Lymphoma/drug therapy , Panobinostat/administration & dosage , Administration, Oral , Adult , Child , Female , Hematologic Neoplasms/blood , Humans , Hypercholesterolemia/blood , Hypercholesterolemia/chemically induced , Leukemia/blood , Lymphoma/blood , Male , Panobinostat/adverse effects , Recurrence
3.
Lancet Oncol ; 19(5): 649-659, 2018 05.
Article in English | MEDLINE | ID: mdl-29650362

ABSTRACT

BACKGROUND: Activating enhancer of zeste homolog 2 (EZH2) mutations or aberrations of the switch/sucrose non-fermentable (SWI/SNF) complex (eg, mutations or deletions of the subunits INI1 or SMARCA4) can lead to aberrant histone methylation, oncogenic transformation, and a proliferative dependency on EZH2 activity. In this first-in-human study, we aimed to investigate the safety, clinical activity, pharmacokinetics, and pharmacodynamics of tazemetostat, a first-in-class selective inhibitor of EZH2. METHODS: We did an open-label, multicentre, dose-escalation, phase 1 study using a 3 + 3 design with planned cohort expansion at the two highest doses below the maximally tolerated dose. The study was done at two centres in France: Institut Gustave Roussy (Villejuif, Val de Marne) and Institut Bergonié (Bordeaux, Gironde). Eligible patients had relapsed or refractory B-cell non-Hodgkin lymphoma or an advanced solid tumour and were older than 18 years, with Eastern Cooperative Oncology Group performance status of 0 or 1, and adequate end-organ function. Tazemetostat was administered orally from 100 mg twice daily to 1600 mg twice daily in 28-day cycles. The primary endpoint was to establish the maximum tolerated dose or recommended phase 2 dose of tazemetostat, as determined by dose-limiting toxicities, laboratory values, and other safety or pharmacokinetic measures in cycle one according to local investigator assessment. Safety was assessed in patients who received at least one dose of tazemetostat; antitumour activity was assessed in the intention-to-treat population. This study is registered with ClinicalTrials.gov, number NCT01897571. The phase 1 part of the study is complete, and phase 2 is ongoing. FINDINGS: Between June 13, 2013, and Sept 21, 2016, 64 patients (21 with B-cell non-Hodgkin lymphoma, and 43 with advanced solid tumours) received doses of tazemetostat. The most common treatment-related adverse events, regardless of attribution, were asthenia (21 [33%] of 64 treatment-related events), anaemia (nine [14%]), anorexia (four [6%]), muscle spasms (nine [14%]), nausea (13 [20%]), and vomiting (six [9%]), usually grade 1 or 2 in severity. A single dose-limiting toxicity of grade 4 thrombocytopenia was identified at the highest dose of 1600 mg twice daily. No treatment-related deaths occurred; seven (11%) patients had non-treatment-related deaths (one at 200 mg twice daily, four at 400 mg twice daily, and two at 1600 mg twice daily). The recommended phase 2 dose was determined to be 800 mg twice daily. Durable objective responses, including complete responses, were observed in eight (38%) of 21 patients with B-cell non-Hodgkin lymphoma and two (5%) of 43 patients with solid tumours. INTERPRETATION: Tazemetostat showed a favourable safety profile and antitumour activity in patients with refractory B-cell non-Hodgkin lymphoma and advanced solid tumours, including epithelioid sarcoma. Further clinical investigation of tazemetostat monotherapy is ongoing in phase 2 studies in adults and a phase 1 study for children, which are currently enrolling patients who have B-cell non-Hodgkin lymphoma and INI1-negative or SMARCA4-negative tumours. FUNDING: Epizyme and Eisai.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzamides/administration & dosage , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Lymphoma, B-Cell/drug therapy , Pyridones/administration & dosage , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Benzamides/adverse effects , Benzamides/pharmacokinetics , Biphenyl Compounds , Dose-Response Relationship, Drug , Enhancer of Zeste Homolog 2 Protein/metabolism , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , France , Humans , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/pathology , Male , Maximum Tolerated Dose , Middle Aged , Morpholines , Pyridones/adverse effects , Pyridones/pharmacokinetics , Time Factors , Treatment Outcome
4.
Pediatr Blood Cancer ; 62(6): 1048-54, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25662999

ABSTRACT

BACKGROUND: FLT3/ITD is associated with poor outcomes in adult and pediatric acute myeloid leukemia (AML). Allogeneic hematopoietic stem cell transplantation (HSCT) can improve cure rates, however relapse is still common. Recent studies demonstrate the activity of FLT3 inhibitors, including sorafenib, in targeting the underlying mutation. PROCEDURE: We conducted a retrospective study of 15 pediatric patients with FLT3/ITD+ AML treated with sorafenib within 18 months after receiving HSCT. Sorafenib was administered either as prophylaxis in patients considered at very high risk for relapse (n = 6) or at the time of disease recurrence (n = 9). RESULTS: Sorafenib was initiated at a median of 100 days post HSCT. Overall, 11/15 (73%) of patients experienced medically significant toxicities. Among patients who experienced toxicity, 6/11 (55%) received treatment at doses above what was later determined to be the maximum tolerated dose of sorafenib for pediatric leukemia. Importantly, sorafenib did not appear to exacerbate graft versus host disease. Our findings suggest that sorafenib may be of particular efficacy in patients with minimal residual disease (MRD); all patients who received sorafenib for MRD immediately prior to transplant or with emergence post-HSCT are alive and remain in complete remission at a median of 48 months post HSCT. CONCLUSIONS: Our case series suggests that sorafenib administration is feasible and tolerable in pediatric FLT3/ITD+ AML patients early post HSCT. Ongoing prospective controlled studies are needed to further define the dosing of sorafenib in the post-HSCT period and to determine the optimal context for this treatment approach.


Subject(s)
Antineoplastic Agents/therapeutic use , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Tandem Repeat Sequences , fms-Like Tyrosine Kinase 3/genetics , Adolescent , Adult , Child , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Neoplasm, Residual , Niacinamide/adverse effects , Niacinamide/therapeutic use , Phenylurea Compounds/adverse effects , Retrospective Studies , Sorafenib
5.
Blood ; 118(2): 243-51, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21562038

ABSTRACT

Children's Cancer Group-1991 selected 2 components from the Children's Cancer Group studies shown to be effective in high-risk acute lymphoblastic leukemia and examined them in children with National Cancer Institute standard-risk acute B-precursor lymphoblastic leukemia. These were (1) vincristine and escalating IV methotrexate (MTX) without leucovorin rescue during the interim maintenance (IM) phases and (2) addition of a second delayed intensification (DI) phase. Eligible patients (n = 2078) were randomly assigned to regimens containing either oral (PO) MTX, PO mercaptopurine, dexamethasone, and vincristine or IV MTX during IM phases, and regimens with either single DI or double DI. Five-year event-free survival (EFS) and overall survival for patients on the PO MTX arms were 88.7% ± 1.4% and 96% ± 0.9% versus 92.6% ± 1.2% and 96.5% ± 0.8% for those on the IV MTX arms (P = .009, P = .66). Five-year EFS and overall survival for patients who received single DI were 90.9% ± 1.3% and 97.1% ± 0.8% versus 90.5% ± 1.3% and 95.4% ± 3.8% for those who received double DI (P = .71, P = .12). No advantage was found for a second DI; however, replacement of PO MTX, PO mercaptopurine, vincristine, and dexamethasone during IM with vincristine and escalating IV MTX improved EFS.


Subject(s)
Methotrexate/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Algorithms , Antineoplastic Agents/administration & dosage , Child , Child, Preschool , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Humans , Infant , Injections, Intravenous , Male , Medical Oncology/methods , Medical Oncology/organization & administration , Neoplasm Staging , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Risk , Societies, Medical
6.
Blood ; 118(23): 6043-9, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21967976

ABSTRACT

The outcomes in children with refractory/relapsed (R/R) acute lymphoblastic leukemia (ALL) are dismal. The efficacy and safety of intravenous clofarabine 40 mg/m(2) per day, cyclophosphamide 440 mg/m(2) per day, and etoposide 100 mg/m(2) per day for 5 consecutive days in pediatric patients with R/R ALL was evaluated in this phase 2 study. The primary endpoint was overall response rate (complete remission [CR] plus CR without platelet recovery [CRp]). Among the 25 patients (median age, 14 years; pre-B cell ALL, 84%; ≥ 2 prior regimens: 84%; refractory to previous regimen: 60%), the overall response rate was 44% (7 CR, 4 CRp) with a 67.3-week median duration or remission censored at last follow-up. Most patients proceeded to alternative therapy, and 10 patients (40%) received hematopoietic stem cell transplantation. Six patients (24%) died because of treatment-related adverse events associated with infection, hepatotoxicity, and/or multiorgan failure. The study protocol was amended to exclude patients with prior hematopoietic stem cell transplantation after 4 of the first 8 patients developed severe hepatotoxicity suggestive of veno-occlusive disease. No additional cases of veno-occlusive disease occurred. The regimen offered encouraging response rates and sustained remission in R/R patients. Future investigation should include exploration of patient selection, dosing, and supportive care. This trial was registered at www.clinicaltrials.gov as #NCT00315705.


Subject(s)
Adenine Nucleotides/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Arabinonucleosides/administration & dosage , Cyclophosphamide/administration & dosage , Etoposide/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adenine Nucleotides/adverse effects , Adolescent , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Arabinonucleosides/adverse effects , Child , Child, Preschool , Clofarabine , Cyclophosphamide/adverse effects , Etoposide/adverse effects , Female , Humans , Infant , Kaplan-Meier Estimate , Male , Nucleoside Transport Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , RNA, Messenger/metabolism , Recurrence , Remission Induction , Risk Factors , Young Adult
7.
Pediatr Blood Cancer ; 57(1): 163-5, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21370437

ABSTRACT

Previous studies to evaluate minimal disease in acute lymphoblastic leukemia (ALL) after treatment have relied on the diagnostic specimen to develop patient-specific analytical probes. The diagnostic specimen is often not available in a tertiary setting; therefore, we evaluated the use of flow cytometry (FCM) using a "difference from normal" approach to detect residual disease prior to myeloablative allogeneic hematopoietic cell transplantation (HCT). Among 116 pediatric patients with ALL who were in morphological remission at time of transplant, we found that those patients who had detectable residual disease by FCM prior to HCT experienced significantly inferior outcome.


Subject(s)
Hematopoietic Stem Cell Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Infant , Male , Neoplasm, Residual , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Retrospective Studies , Survival Rate , Transplantation, Homologous
9.
Acad Med ; 80(11): 1012-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16249299

ABSTRACT

The translation of hypothesis-driven research laboratory findings about basic disease mechanisms into clinically useful tests or therapies, particularly in pediatric diseases, is time-consuming, expensive, and not well supported by traditional research grant mechanisms. Accordingly, the development of new drugs and clinical assays has typically been largely the domain of the pharmaceutical industry. Aside from partnering with for-profit companies, academic health centers are challenged to find ways to actively engage in biomedical research to bridge the gap between basic and clinical research. The Translational Research Initiative (TRI) at Cincinnati Children's Hospital Medical Center was launched in 2001 with the mission to build an institutional infrastructure for promoting and facilitating the clinical implementation of investigator-initiated basic research. The TRI's goals are to provide grant support for proposals that are translational in nature and that address serious diagnostic or therapeutic deficiencies in pediatric illnesses; to create and support specialized research cores and a specialized office that provides support for research protocol development and regulatory affairs; and to organize educational opportunities focused on bridging communication between basic and clinical scientists and encouraging multidisciplinary interactions. The authors describe the program structure and provide an interim outcome report as measured by extramural funding obtained, Investigational New Drug applications filed, manuscripts published, clinical trials launched, and educational initiatives created. The broad success of this program suggests that it might serve as a model for other academic health centers in promoting and conducting translational research.


Subject(s)
Academic Medical Centers/organization & administration , Biomedical Research/methods , Research Support as Topic/organization & administration , Academic Medical Centers/economics , Biomedical Research/economics , Child , Diffusion of Innovation , Financing, Organized/standards , Hospitals, Pediatric , Humans , Investigational New Drug Application , National Institutes of Health (U.S.) , Ohio , Organizational Objectives , Program Development , Research Personnel/education , Research Support as Topic/economics , Research Support as Topic/standards , United States
10.
Clin Cancer Res ; 10(16): 5335-41, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15328169

ABSTRACT

PURPOSE: Asparaginase therapy is an important component in the treatment of children with acute lymphoblastic leukemia. Polyethylene glycol-conjugated asparaginase (PEG-ASNase) has significant pharmacological advantages over native Escherichia coli asparaginase. We investigated the pharmacokinetics of PEG-ASNase, presence of antibodies to PEG-ASNase, and concentrations of asparagine in serum and cerebrospinal fluid (CSF) in combination chemotherapy for relapsed pediatric acute lymphoblastic leukemia. EXPERIMENTAL DESIGN: Twenty-eight pediatric patients with relapsed medullary (n = 16) and extramedullary (n = 11) acute lymphoblastic leukemia were enrolled at three pediatric institutions and had at least two serum and CSF samples obtained for analysis. Patients received induction therapy (including PEG-ASNase 2500 IU/m2 intramuscularly weekly on days 2, 9, 16, and 23) and intensification therapy (including PEG-ASNase 2500 IU/m2 intramuscularly once on day 7). Serum samples were obtained weekly during induction and intensification. CSF samples were obtained during therapeutic lumbar punctures during induction and intensification. RESULTS: Weekly PEG-ASNase therapy resulted in PEG-ASNase activity of >0.1 IU/ml in 91-100% of patients throughout induction. During intensification, PEG-ASNase on day 7 resulted in PEG-ASNase activity >0.1 IU/ml in 94% and 80% of patients on days 14 and 21, respectively. Serum and CSF asparagine depletion was observed and maintained during induction and intensification in the majority of samples. PEG-ASNase antibody was observed in only 3 patients. CONCLUSIONS: Intensive PEG-ASNase therapy in the treatment of relapsed acute lymphoblastic leukemia reliably results in high-level serum PEG-ASNase activity, and asparagine depletion in serum and CSF is usually achieved. Incorporation of intensive PEG-ASNase in future trials for recurrent acute lymphoblastic leukemia is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asparaginase/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Asparaginase/therapeutic use , Asparaginase/toxicity , Asparagine/blood , Asparagine/cerebrospinal fluid , Child , Child, Preschool , Delayed-Action Preparations , Female , Glutamine/blood , Humans , Immunophenotyping , Male , Polyethylene Glycols/therapeutic use , Polyethylene Glycols/toxicity , Recurrence
11.
J Pediatric Infect Dis Soc ; 3(4): 286-93, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25419459

ABSTRACT

BACKGROUND: The clinical presentation and management of human metapneumovirus (hMPV) infections in immunocompromised children is not well understood. METHODS: We performed a retrospective evaluation of pediatric patients with laboratory-confirmed hMPV infections and underlying hematologic malignancy, solid tumors, solid organ transplant, rheumatologic disease, and/or receipt of chronic immunosuppressants. Data were analyzed using t tests and Fisher's exact tests. RESULTS: Overall, 55 patients (median age: 5 years; range: 5 months-19 years) with hMPV infection documented between 2006 and 2010 were identified, including 24 (44%) with hematologic malignancy, 9 (16%) undergoing hematopoietic stem cell transplant, 9 (16%) with solid tumors, and 8 (15%) with solid organ transplants. Three (5%) presented with fever alone, 35 (64%) presented with upper respiratory tract infections, and 16 (29%) presented with lower respiratory tract infections (LRTI). Twelve (23%) patients required intensive care unit admission and/or supplemental oxygen ≥28% FiO2. Those with severe disease were more likely to be neutropenic (P = .02), but otherwise did not differ by age (P = .27), hematopoietic stem cell transplant recipient status (P = .19), or presence of lymphopenia (P = .09). Nine (16%) patients received treatment with ribavirin, intravenous immunoglobulin, or both. Three children (5%) died of hMPV pneumonia. CONCLUSIONS: Immunocompromised pediatric patients with hMPV infection have high rates of LRTI and mortality. The benefits of treatment with ribavirin and intravenous immunoglobulin in this patient population require further evaluation.

13.
Arch Pediatr Adolesc Med ; 164(3): 214-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20194252

ABSTRACT

OBJECTIVE: To determine whether clinical trial enrollment by itself is associated with improved outcome. DESIGN: Retrospective cohort study. SETTING: Seattle Children's Hospital from 1997 to 2005. PARTICIPANTS: Data were drawn from 322 patients with newly diagnosed acute lymphoblastic leukemia. Main Exposure Enrollment in a Children's Oncology Group or Children's Cancer Group clinical trial. MAIN OUTCOME MEASURES: (1) Demographic variables associated with trial participation. (2) Event-free survival, which was defined as the time from initial diagnosis to either leukemia recurrence or death from any cause. RESULTS: No outcome advantage was found for participants in a clinical trial compared with nonparticipants. Additionally, there were not demographic factors associated with increased clinical trial participation. CONCLUSIONS: Clinical trial participation does not, by itself, lead to improved outcome for pediatric patients with acute lymphoblastic leukemia in the current era. Discussions about participation in a clinical trial should focus on improvement of future therapy, not the direct benefit of the research participant.


Subject(s)
Clinical Trials as Topic , Health Knowledge, Attitudes, Practice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Retrospective Studies , Survival Analysis , Treatment Outcome , Washington/epidemiology , Young Adult
14.
Pediatr Blood Cancer ; 43(5): 571-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15382275

ABSTRACT

BACKGROUND: Approximately 25% of children newly diagnosed with acute lymphoblastic leukemia (ALL) will eventually experience leukemic relapse, with bone marrow being the most common site of recurrence. The ability to achieve a durable second remission is complicated by toxicity and resistant disease. We report a novel combination of chemotherapy for relapsed pediatric ALL. PROCEDURE: Thirty pediatric patients with relapsed medullary (n = 18) and extra-medullary (n = 12) ALL were enrolled at three pediatric institutions. Following receipt of induction and the first Block A and Block B of intensification, each patient was evaluated for toxicity, efficacy in achieving remission, and long-term survival. Additionally, minimal residual disease (MRD) detection by multidimensional flow cytometry (MDF) was performed. RESULTS: During induction, the major non-hematopoeitic toxicities were mucositis (30% of patients) and bacteremia (50% of patients). Two patients (7%) died of toxicity during induction. Toxicity during intensification Block 1A and 1B was markedly reduced. Eight-nine percent of patients with marrow disease achieved a remission following induction and intensification. The event-free survival (EFS) for all patients at 2 and 4 years were 60% (95% CI: 42-78%) and 49% (95% CI: 30-68%), respectively. CONCLUSIONS: This regimen for patients with relapsed ALL was successful in achieving a second remission for the majority of patients with acceptable toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Neoplasms/drug therapy , Bone Marrow Neoplasms/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Administration, Oral , Asparaginase/administration & dosage , Bacteremia/chemically induced , Child , Child, Preschool , Cytarabine/administration & dosage , Dexamethasone/administration & dosage , Etoposide/administration & dosage , Female , Flow Cytometry , Humans , Idarubicin/administration & dosage , Ifosfamide/administration & dosage , Infant , Infusions, Intravenous , Leucovorin/administration & dosage , Male , Mesna/administration & dosage , Methotrexate/administration & dosage , Mouth Mucosa/pathology , Polyethylene Glycols/administration & dosage , Recurrence , Stomatitis/chemically induced , Thioguanine/administration & dosage , Treatment Outcome , Vincristine/administration & dosage
15.
Cancer ; 95(6): 1354-65, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12216105

ABSTRACT

BACKGROUND: To increase the dose intensity (DI) of chemotherapy for pediatric patients with metastatic sarcomas, including the Ewing sarcoma family of tumors (ESFT) and rhabdomyosarcoma (RMS), the authors tested the feasibility of an intensive regimen supported by granulocyte-colony stimulating factor (G-CSF) and peripheral blood stem cells (PBSC). METHODS: Twenty-three children and adolescents with metastatic sarcomas received vincristine, doxorubicin, cyclophosphamide, ifosfamide, sodium mercaptoethanesulfonate (mensa), and etoposide (VACIME) chemotherapy, consisting of 8 courses of vincristine 2 mg/m(2) on Day 0, doxorubicin 37.5 mg/m(2) per day on Days 0-1, cyclophosphamide 360 mg/m(2) per day on Days 0-4, ifosfamide 1800 mg/m(2) per day on Days 0-4, mesna 2400 mg/m(2) per day, and etoposide 100 mg/m(2) per day on Days 0-4. Doxorubicin was omitted in Courses 7 and 8. G-CSF was given after each course of therapy. Courses of therapy were repeated every 21 days or as soon as hematopoietic recovery permitted. PBSC were collected twice: first, after Course 2 (infused after Courses 3 and 4) and, second, after Course 4 (infused after Courses 5 and 6). Surgical resection followed Course 6, and radiotherapy followed Course 8. RESULTS: PBSC collections were adequate in 91% of all harvests. The mean DI was 82% (standard deviation, 14%) of the intended DI, which was greater than historic data without PBSC support. Seventeen patients (74%) achieved a complete response (CR), 12 patients with chemotherapy alone and 5 more patients after undergoing surgical resection. Fifteen patients developed progressive disease, with a 2-year event free survival (EFS) rate of 39% (95% confidence interval, 19-59%). Hematopoietic toxicity was severe and cumulative, although it was less than that seen previously without PBSC support. CONCLUSIONS: PBSC-supported multicycle chemotherapy is a feasible method to increase chemotherapy DI for pediatric patients with metastatic sarcomas. Although the CR rate compared favorably with previously reported response rates, the 2-year EFS rate was similar to that achieved with other intensive regimens.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cyclophosphamide/adverse effects , Doxorubicin/adverse effects , Etoposide/adverse effects , Hematopoietic Stem Cell Transplantation , Ifosfamide/adverse effects , Mesna/adverse effects , Sarcoma/therapy , Soft Tissue Neoplasms/therapy , Vincristine/adverse effects , Adolescent , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Child , Child, Preschool , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Etoposide/administration & dosage , Feasibility Studies , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Ifosfamide/administration & dosage , Infant , Male , Mesna/administration & dosage , Neoplasm Metastasis , Remission Induction , Sarcoma, Ewing/therapy , Treatment Outcome , Vincristine/administration & dosage
16.
J Allergy Clin Immunol ; 113(1): 115-9, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14699394

ABSTRACT

BACKGROUND: IL-5 is a cytokine critically involved in regulating several aspects of eosinophils including their production, activation, and tissue recruitment. As such, IL-5 may be involved in the pathogenesis of hypereosinophilic syndromes, a group of poorly treated diverse disorders characterized by sustained peripheral blood and/or tissue eosinophilia. OBJECTIVE: We aimed to assess the safety and efficacy of a humanized blocking monoclonal antibody against IL-5 (mepolizumab) in patients with several forms of hyper-eosinophilic syndromes. METHODS: We performed an open-label trial of anti-IL-5 in which 3 intravenous doses (10 mg/kg, maximum 750 mg) were administered at 4-week intervals to 4 patients with hypereosinophilic syndromes (defined by peripheral blood and/or tissue eosinophilia). The effects of treatment on safety, eosinophil levels (in peripheral blood and/or diseased tissue), pulmonary function, and quality of life were measured over a 28-week period. RESULTS: Anti-IL-5 was well tolerated in all patients and lowered peripheral blood eosinophil counts despite ongoing systemic glucocorticoid therapy. The decline in circulating eosinophil counts was sustained for at least 12 weeks after the last dose of anti-IL-5. In addition, anti-IL-5 improved clinical and quality of life measurements. In one patient with striking tissue eosinophilia (eosinophilic esophagitis), anti-IL-5 resulted in a 10-fold reduction in tissue eosinophil levels. CONCLUSIONS: These results suggest that anti-IL-5 is safe, effective in lowering eosinophil levels, and has potential glucocorticoid-sparing effects in patients with a variety of hyper-eosinophilic syndromes. As such, anti-IL-5 may have significant therapeutic potential for hypereosinophilic syndromes.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hypereosinophilic Syndrome/drug therapy , Interleukin-5/immunology , Adolescent , Adult , Antibodies, Monoclonal, Humanized , Drug Therapy, Combination , Eosinophils/immunology , Female , Glucocorticoids/therapeutic use , Humans , Hypereosinophilic Syndrome/immunology , Leukocyte Count , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL