Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Sci Transl Med ; 15(687): eabn2110, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36921036

ABSTRACT

Among drug-induced adverse events, pancreatitis is life-threatening and results in substantial morbidity. A prototype example is the pancreatitis caused by asparaginase, a crucial drug used to treat acute lymphoblastic leukemia (ALL). Here, we used a systems approach to identify the factors affecting asparaginase-associated pancreatitis (AAP). Connectivity Map analysis of the transcriptomic data showed that asparaginase-induced gene signatures were potentially reversed by retinoids (vitamin A and its analogs). Analysis of a large electronic health record database (TriNetX) and the U.S. Federal Drug Administration Adverse Events Reporting System demonstrated a reduction in AAP risk with concomitant exposure to vitamin A. Furthermore, we performed a global metabolomic screening of plasma samples from 24 individuals with ALL who developed pancreatitis (cases) and 26 individuals with ALL who did not develop pancreatitis (controls), before and after a single exposure to asparaginase. Screening from this discovery cohort revealed that plasma carotenoids were lower in the cases than in controls. This finding was validated in a larger external cohort. A 30-day dietary recall showed that the cases received less dietary vitamin A than the controls did. In mice, asparaginase administration alone was sufficient to reduce circulating and hepatic retinol. Based on these data, we propose that circulating retinoids protect against pancreatic inflammation and that asparaginase reduces circulating retinoids. Moreover, we show that AAP is more likely to develop with reduced dietary vitamin A intake. The systems approach taken for AAP provides an impetus to examine the role of dietary vitamin A supplementation in preventing or treating AAP.


Subject(s)
Antineoplastic Agents , Pancreatitis , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Mice , Asparaginase/adverse effects , Retinoids/adverse effects , Vitamin A/therapeutic use , Pancreatitis/chemically induced , Pancreatitis/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Systems Analysis , Antineoplastic Agents/adverse effects
3.
Cancer Med ; 10(21): 7551-7560, 2021 11.
Article in English | MEDLINE | ID: mdl-34528411

ABSTRACT

BACKGROUND: Pegaspargase (PEG-ASP) is an integral component of therapy for acute lymphoblastic leukemia (ALL) but is associated with hepatotoxicity that may delay or limit future therapy. Obese and adolescent and young adult (AYA) patients are at high risk. Levocarnitine has been described as potentially beneficial for the treatment or prevention of PEG-ASP-associated hepatotoxicity. METHODS: We collected data for patients age ≥10 years who received levocarnitine during induction therapy for ALL, compared to a similar patient cohort who did not receive levocarnitine. The primary endpoint was conjugated bilirubin (c.bili) >3 mg/dl. Secondary endpoints were transaminases >10× the upper limit of normal and any Grade ≥3 hepatotoxicity. RESULTS: Fifty-two patients received levocarnitine for prophylaxis (n = 29) or rescue (n = 32) of hepatotoxicity. Compared to 109 patients without levocarnitine, more patients receiving levocarnitine were obese and/or older and had significantly higher values for some hepatotoxicity markers at diagnosis and after PEG-ASP. Levocarnitine regimens varied widely; no adverse effects of levocarnitine were identified. Obesity and AYA status were associated with an increased risk of conjugated hyperbilirubinemia and severe transaminitis. Multivariable analysis identified a protective effect of levocarnitine on the development of c.bili >3 mg/dl (OR 0.12, p = 0.029). There was no difference between groups in CTCAE Grade ≥3 hepatotoxicity. C.bili >3 mg/dl during induction was associated with lower event-free survival. CONCLUSIONS: This real-world data on levocarnitine supplementation during ALL induction highlights the risk of PEG-ASP-associated hepatotoxicity in obese and AYA patients, and hepatotoxicity's potential impact on survival. Levocarnitine supplementation may be protective, but prospective studies are needed to confirm these findings.


Subject(s)
Antineoplastic Agents/adverse effects , Asparaginase/adverse effects , Carnitine/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/etiology , Polyethylene Glycols/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Chemical and Drug Induced Liver Injury/prevention & control , Child , Female , Humans , Induction Chemotherapy , Male , Pediatric Obesity/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Survival Analysis , Young Adult
4.
Cancer Chemother Pharmacol ; 87(6): 817-826, 2021 06.
Article in English | MEDLINE | ID: mdl-33677674

ABSTRACT

PURPOSE: Asparaginases, key agents in treatment of acute lymphoblastic leukemia (ALL), are associated with venous thromboembolism (VTE). While risks of short-acting asparaginase-related VTE is well-known, we studied VTE incidence and risk factors in adult ALL patients treated with and without long-acting pegylated asparaginase (PegA). METHODS: Single-center, retrospective analysis of 89 ALL patients treated with (n = 61) or without (n = 28) PegA at Greenebaum Comprehensive Cancer Center. Reviewed patient and disease characteristics, treatment, and VTE incidence. RESULTS: VTE during treatment occurred in 31 patients (35%), and was associated with PegA (p = 0.001) and Philadelphia chromosome negativity (p = 0.002). Among PegA recipients, VTE was associated with a significantly higher mean body mass index (BMI) of 31.3 kg/m2 (p = 0.037), and was more common with pre-T/T cell compared to pre-B/B cell ALL (68.2% vs. 33.3%, p = 0.009). Antithrombin-III (ATIII) levels were measured for 26 patients; 16 (61.5%) were < 50%. Of those, 8 (50%) experienced VTE, while 3 of 10 (30%) patients with ATIII levels ≥ 50% experienced VTE. VTE occurred in 7 of 13 (54%) of patients who received ATIII repletion. There was a trend toward a higher incidence of VTE in the PegA group among patients with non-O compared to O blood type (55.9% vs. 33.3%, p = 0.079) as well as those with a higher hemoglobin at diagnosis (9.3 vs 8.1 g/dL, p = 0.056). CONCLUSION: This study confirms PegA as a risk factor for VTE in patients with ALL. Risk factors among those receiving PegA include higher BMI and pre-T/T cell ALL. ATIII repletion was not shown to be protective against VTE. There was a higher incidence of VTE in patients who received PegA with non-O compared to O blood type, but the precise correlation is uncertain.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Asparaginase/adverse effects , Asparaginase/pharmacology , Escherichia coli/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Venous Thromboembolism/chemically induced , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Antithrombin III/metabolism , B-Lymphocytes/drug effects , Body Mass Index , Female , Humans , Incidence , Male , Middle Aged , Retrospective Studies , Risk Factors , T-Lymphocytes/drug effects , Young Adult
5.
J Clin Oncol ; 38(28): 3282-3293, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32813610

ABSTRACT

PURPOSE: Nelarabine is effective in inducing remission in patients with relapsed and refractory T-cell acute lymphoblastic leukemia (T-ALL) but has not been fully evaluated in those with newly diagnosed disease. PATIENTS AND METHODS: From 2007 to 2014, Children's Oncology Group trial AALL0434 (ClinicalTrials.gov identifier: NCT00408005) enrolled 1,562 evaluable patients with T-ALL age 1-31 years who received the augmented Berlin-Frankfurt-Muenster (ABFM) regimen with a 2 × 2 pseudo-factorial randomization to receive escalating-dose methotrexate (MTX) without leucovorin rescue plus pegaspargase (C-MTX) or high-dose MTX (HDMTX) with leucovorin rescue. Intermediate- and high-risk patients were also randomly assigned after induction to receive or not receive six 5-day courses of nelarabine that was incorporated into ABFM. Patients who experienced induction failure were nonrandomly assigned to HDMTX plus nelarabine. Patients with overt CNS disease (CNS3; ≥ 5 WBCs/µL with blasts) received HDMTX and were randomly assigned to receive or not receive nelarabine. All patients, except those with low-risk disease, received cranial irradiation. RESULTS: The 5-year event-free and overall survival rates were 83.7% ± 1.1% and 89.5% ± 0.9%, respectively. The 5-year disease-free survival (DFS) rates for patients with T-ALL randomly assigned to nelarabine (n = 323) and no nelarabine (n = 336) were 88.2% ± 2.4% and 82.1% ± 2.7%, respectively (P = .029). Differences between DFS in a four-arm comparison were significant (P = .01), with no interactions between the MTX and nelarabine randomizations (P = .41). Patients treated with the best-performing arm, C-MTX plus nelarabine, had a 5-year DFS of 91% (n = 147). Patients who received nelarabine had significantly fewer isolated and combined CNS relapses compared with patients who did not receive nelarabine (1.3% ± 0.63% v 6.9% ± 1.4%, respectively; P = .0001). Toxicities, including neurotoxicity, were acceptable and similar between all four arms. CONCLUSION: The addition of nelarabine to ABFM therapy improved DFS for children and young adults with newly diagnosed T-ALL without increased toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Arabinonucleosides/administration & dosage , Arabinonucleosides/adverse effects , Asparaginase/administration & dosage , Asparaginase/adverse effects , Child , Cohort Studies , Disease-Free Survival , Female , Humans , Leucovorin/administration & dosage , Leucovorin/adverse effects , Male , Methotrexate/administration & dosage , Methotrexate/adverse effects , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Prednisone/administration & dosage , Prednisone/adverse effects , Treatment Outcome
6.
Leuk Lymphoma ; 61(9): 2200-2207, 2020 09.
Article in English | MEDLINE | ID: mdl-32482107

ABSTRACT

Pegaspargase (PEG) increases venous thromboembolism (VTE) in acute lymphoblastic leukemia (ALL) potentially due to depletion of anticoagulation factors, including antithrombin (AT). The benefit and cost of AT supplementation in adults is unclear. We aimed to characterize VTE incidence and risk factors following AT and determine the characteristics and costs of supplementation. Fifty-three adults received PEG and AT. VTE occurred in 21% (grade ≥3 8%). T cell ALL and patients receiving prednisone during induction were at highest risk. Repeat AT levels post supplementation were subtherapeutic forty-four percent of the time. A median of 18 days elapsed between PEG and two sequential therapeutic AT levels despite supplementation. Patients received a median of 2 AT doses per PEG dose at a median cost of $11,145. VTE remains common in adults despite AT supplementation. More aggressive AT supplementation may reduce VTE but warrant prospective evaluation given the significant cost.


Subject(s)
Venous Thromboembolism , Adult , Antithrombins/adverse effects , Asparaginase/adverse effects , Dietary Supplements , Humans , Polyethylene Glycols , Prospective Studies , Risk Factors , Venous Thromboembolism/drug therapy , Venous Thromboembolism/epidemiology , Venous Thromboembolism/etiology
7.
J Oncol Pharm Pract ; 26(1): 74-92, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30917738

ABSTRACT

The incorporation of L-asparaginase and pegylated asparaginase into pediatric-inspired regimens has conferred a survival advantage in treatment of adults with acute lymphoblastic leukemia. Use of asparaginase products requires careful prevention, monitoring, and management of adverse effects including hypersensitivity, hepatotoxicity, pancreatitis, coagulopathy, and thrombosis. Currently, there is limited published literature to offer guidance on management of these toxicities. At the University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, a standard of practice guideline was created to prevent and manage asparaginase-related adverse events. By sharing our long-term experience with asparaginase products and clinical management of asparaginase-induced toxicities, this article aims to improve patient safety and optimize treatment outcomes.


Subject(s)
Antineoplastic Agents/administration & dosage , Asparaginase/administration & dosage , Cancer Care Facilities/standards , Disease Management , Drug Monitoring/standards , Polyethylene Glycols/administration & dosage , Practice Guidelines as Topic/standards , Adult , Antineoplastic Agents/adverse effects , Asparaginase/adverse effects , Child, Preschool , Dose-Response Relationship, Drug , Drug Monitoring/methods , Humans , Polyethylene Glycols/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/epidemiology , Thrombosis/chemically induced , Thrombosis/epidemiology , Thrombosis/prevention & control , Treatment Outcome
8.
PLoS One ; 14(5): e0216328, 2019.
Article in English | MEDLINE | ID: mdl-31059548

ABSTRACT

INTRODUCTION: Combination therapy for acute lymphoblastic leukemia (ALL) is highly effective but results in significant toxicity including osteonecrosis. Asparaginase is known to potentiate both the antileukemic and osteonecrosis-inducing effects of dexamethasone. The schedule of dexamethasone alters osteonecrosis risk. However, the effects of the interaction with asparaginase are unknown when dexamethasone is given on a discontinuous schedule. METHODS: Using the murine model of osteonecrosis, we compared the frequency of osteonecrosis in mice receiving discontinuous dexamethasone (3.5 days/ week) with mice receiving asparaginase and discontinuous dexamethasone. We then tested the effect on antileukemic efficacy using six pediatric ALL xenografts. RESULTS: The addition of asparaginase to discontinuous dexamethasone did not alter the rate of osteonecrosis compared to dexamethasone alone (7/35 in dexamethasone with asparaginase combination vs. 10/36 in dexamethasone alone, p = 0.62) despite increasing steady-state plasma dexamethasone levels (103.9 nM vs. 33.4 nM, p = 9.2x10-7). Combination therapy with asparaginase and dexamethasone demonstrated synergistic antileukemic effects across all six xenografts studied. CONCLUSIONS: When discontinuous dexamethasone was given, its anti-leukemic activity synergized with asparaginase but the osteonecrosis-worsening effects of asparaginase (above dexamethasone alone) were not observed. Thus, there is a favorable drug interaction (unchanged toxicity, synergistic efficacy) between discontinuous dexamethasone and asparaginase.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asparaginase/administration & dosage , Dexamethasone/administration & dosage , Osteonecrosis/chemically induced , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Asparaginase/adverse effects , Dexamethasone/adverse effects , Drug Evaluation, Preclinical , Drug Interactions , Heterografts , Humans , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications
9.
Chemotherapy ; 64(4): 210-214, 2019.
Article in English | MEDLINE | ID: mdl-32045924

ABSTRACT

BACKGROUND: Pegylated asparaginase may induce prolonged hypertriglyceridemia. To date, there is no standard management of this complication. Here, we present a case report of pegylated asparaginase-induced hypertriglyceridemia and hepatotoxicity successfully treated with continuous intravenous infusion of insulin and heparin. CASE PRESENTATION: A 51-year-old male patient with lymphoid blast crisis of chronic myelogenous leukemia was treated with pegylated asparaginase. The patient developed severe hypertriglyceridemia. Supportive therapy with low-fat diet, fibric acids, and omega-3 fatty acids was not successful, and later, the patient developed high-grade hepatotoxicity. Like hypertriglyceridemia-induced pancreatitis, continuous intravenous infusion of insulin and heparin was initiated. The level of triglyceride and cholesterol decreased rapidly within 4 days. CONCLUSION: In case of severe pegylated asparaginase-induced hypertriglyceridemia, continuous intravenous infusion of insulin and heparin can reduce rapidly and safely the triglyceride level. Controlled trials are needed to address this important issue.


Subject(s)
Asparaginase/adverse effects , Heparin/therapeutic use , Hypertriglyceridemia/drug therapy , Insulin/therapeutic use , Polyethylene Glycols/adverse effects , Asparaginase/therapeutic use , Cholesterol/blood , Diet, Fat-Restricted , Humans , Hypertriglyceridemia/etiology , Infusions, Intravenous , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Male , Middle Aged , Pancreatitis/etiology , Polyethylene Glycols/therapeutic use , Triglycerides/blood
10.
Cancer Med ; 7(1): 3-12, 2018 01.
Article in English | MEDLINE | ID: mdl-29274118

ABSTRACT

The Children's Oncology Group (COG) develops and implements multi-institutional clinical trials with the primary goal of assessing the efficacy and safety profile of treatment regimens for various pediatric cancers. However, the monetary costs of treatment regimens are not measured. AALL0232 was a COG randomized phase III trial for children with acute lymphoblastic leukemia that found that dexamethasone (DEX) was a more effective glucocorticoid than prednisone (PRED) in patients younger than 10 years, but PRED was equally effective and less toxic in older patients. In addition, high-dose methotrexate (HD-MTX) led to better survival than escalating doses of methotrexate (C-MTX). Cost data from the Pediatric Health Information System database were merged with clinical data from the COG AALL0232 trial. Total and component costs were compared between treatment arms and across hospitals. Inpatient costs were higher in the HD-MTX and DEX arms when compared to the C-MTX and PRED arms at the end of therapy. There was no difference in cost between these arms at last follow-up. Considerable variation in total costs existed across centers to deliver the same therapy that was driven by differences in inpatient days and pharmacy costs. The more effective regimens were found to be more expensive during therapy but were ultimately cost-neutral in longer term follow-up. The variations in cost across centers suggest an opportunity to standardize resource utilization for patients receiving similar therapies, which could translate into reduced healthcare expenditures.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/economics , Drug Costs , Health Expenditures , Hospital Costs , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Age Factors , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Asparaginase/administration & dosage , Asparaginase/adverse effects , Asparaginase/economics , Child , Child, Preschool , Cost-Benefit Analysis , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Dexamethasone/economics , Disease-Free Survival , Drug Administration Schedule , Female , Follow-Up Studies , Hospitalization/economics , Humans , Infant , Infant, Newborn , Leucovorin/administration & dosage , Leucovorin/adverse effects , Leucovorin/economics , Male , Methotrexate/administration & dosage , Methotrexate/adverse effects , Methotrexate/economics , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Polyethylene Glycols/economics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/economics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prednisone/administration & dosage , Prednisone/adverse effects , Prednisone/economics , Young Adult
11.
Medicine (Baltimore) ; 96(46): e8715, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29145310

ABSTRACT

RATIONALE: Pegaspargase has been used in the treatment of acute lymphoblastic leukemia with promising results. However, it has also been associated with several potentially serious complications, including thrombosis. Pegaspargase-induced cerebral venous thrombosis and bone marrow necrosis are very rare. PATIENT CONCERNS: A 50-year-old female developed headache, weakness of the right lower extremity, fever, and bone pain after chemotherapy including pegaspargase for the treatment of acute lymphoblastic leukemia. DIAGNOSES: Her imaging studies and bone marrow examinations were compatible with cerebral venous thrombosis and bone marrow necrosis. INTERVENTIONS: The patient received anticoagulation therapy with rivaroxaban. OUTCOMES: After treatment with rivaroxaban, she had a good outcome without major or minor bleeding. LESSONS: Clinicians should be aware of the very rare but possible induction of bone marrow necrosis during pegaspargase treatment when there is necrosis in other organs. Because of its greater safety and convenience, rivaroxaban gains popularity over traditional anticoagulant drugs.


Subject(s)
Antineoplastic Agents/adverse effects , Asparaginase/adverse effects , Bone Marrow/pathology , Polyethylene Glycols/adverse effects , Rivaroxaban/therapeutic use , Sinus Thrombosis, Intracranial/chemically induced , Sinus Thrombosis, Intracranial/drug therapy , Female , Humans , Middle Aged , Necrosis/chemically induced , Necrosis/drug therapy , Remission Induction
13.
Onkologie ; 35(10): 596-8, 2012.
Article in English | MEDLINE | ID: mdl-23038232

ABSTRACT

BACKGROUND: L-Asparaginase (L-Asp) may induce hypertriglyceridemia; however, this has been mainly observed among pediatric patients. Treatment for L-Asp-induced hypertriglyceridemia is not standardized, ranging from fasting and diet restriction to the invasive plasmapheresis procedure. CASE REPORT: We describe a 53-year-old male patient who presented with L-Asp-induced severe hypertriglyceridemia. He was receiving L-Asp as part of his chemotherapy regimen for natural killer T-cell lymphoma. After the 20th dose, his serum triglyceride level was 3,552 mg/dl, with a total cholesterol of 418 mg/dl. Despite the high triglyceride, the patient did not present with acute pancreatitis symptoms. Treatment comprising fasting, fenofibrate, and omega-3 fatty acids was initiated. Triglyceride levels dropped rapidly to 1,000 mg/dl within 2 days, and to 268 mg/dl after 10 days. The chemotherapy regimen was subsequently switched to exclude L-Asp. CONCLUSION: L-Asp-induced severe hypertriglyceridemia may occur in adults and may be conservatively managed with fasting, fibrates, and omega-3 fatty acids. Plasmapheresis or continuous insulin infusion may be used for symptomatic patients with high triglyceride levels. Lipidlowering agents should be continued for patients previously treated for hyperlipidemia. Regular monitoring of lipid levels for patients receiving L-Asp is important, especially for those with a prior history of dyslipidemia. Re-challenge with L-Asp can be undertaken on an individual basis.


Subject(s)
Asparaginase/adverse effects , Hypertriglyceridemia/chemically induced , Hypertriglyceridemia/therapy , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Asparaginase/therapeutic use , Humans , Lymphoma, T-Cell/drug therapy , Male , Middle Aged , Treatment Outcome
14.
Farm. hosp ; 36(3): 148-158, mayo-jun. 2012. tab
Article in Spanish | IBECS | ID: ibc-107829

ABSTRACT

Objetivo Revisar las características y el manejo de las reacciones de hipersensibilidad causadas por agentes antineoplásicos. Método Se realizó una búsqueda bibliográfica en las bases de datos Pubmed y EMBASE de los últimos 10 años. Resultados Casi todos los quimioterápicos tienen potencial para causar una reacción de hipersensibilidad, pero determinados grupos han sido asociados con un mayor riesgo, como los derivados del platino, los taxanos, las asparraginasas, los anticuerpos monoclonales y las epipodofilotoxinas. Las manifestaciones clínicas de estas reacciones son variables e impredecibles incluyendo síntomas cutáneos, respiratorios, cardiacos y gastrointestinales. El mecanismo asociado con su desarrollo aún no se conoce en su totalidad. El diagnóstico se basa en los signos y síntomas que desarrolle el paciente y en la realización de pruebas cutáneas. El manejo de los pacientes que sufran una reacción de hipersensibilidad a un quimioterápico variará según el grado de severidad de la reacción, de la necesidad de continuar con el tratamiento y de las alternativas terapéuticas disponibles. Conclusiones Al producirse un incremento progresivo en la utilización de los agentes quimioterápicos, se puede esperar un aumento de la incidencia de las reacciones de hipersensibilidad. Los protocolos de desensibilización destacan como una alternativa que nos van a permitir reintroducir en la terapia del paciente el agente causal de la reacción de hipersensibilidad. Su utilización debe valorarse individualmente sopesando los beneficios y los riesgos (AU)


Objective To review the characteristics and management of hypersensitivity reactions caused by antineoplastic agents. Method We conducted a search in the Pubmed and EMBASE databases for the last 10 years. Results Almost all chemotherapeutic agents have the potential to cause hypersensitivity reactions, but some groups have been associated with increased risk, such as platinum compounds, taxanes, asparaginase, monoclonal antibodies and epipodophyllotoxins. The clinical manifestations of these reactions are variable and unpredictable, including symptoms affecting the skin and the pulmonary, cardiac and gastrointestinal systems. The mechanism associated with their development is not yet fully understood. Diagnosis is based on patients’ signs and symptoms and skin testing. The management of patients who suffer a hypersensitivity reaction to a chemotherapeutic agent varies with the severity of the reaction, the need to continue treatment, and the availability of alternative therapies. Conclusions Due to a progressive increase in the use of chemotherapeutic agents an increased incidence of hypersensitivity reactions is to be expected. Desensitisation protocols are a noteworthy alternative that make it possible to re-initiate patients’ therapy with the causative agent of the hypersensitivity reaction. Their use should be assessed individually, weighing risks and benefits (AU)


Subject(s)
Humans , Antineoplastic Agents/adverse effects , Drug Hypersensitivity/epidemiology , Platinum Compounds/adverse effects , Asparaginase/adverse effects , Taxoids/adverse effects , Desensitization, Immunologic , Neoplasms/drug therapy
15.
Farm Hosp ; 36(3): 148-58, 2012.
Article in English, Spanish | MEDLINE | ID: mdl-22484106

ABSTRACT

OBJECTIVE: To review the characteristics and management of hypersensitivity reactions caused by antineoplastic agents. METHOD: We conducted a search in the Pubmed and EMBASE databases for the last 10 years. RESULTS: Almost all chemotherapeutic agents have the potential to cause hypersensitivity reactions, but some groups have been associated with increased risk, such as platinum compounds, taxanes, asparaginase, monoclonal antibodies and epipodophyllotoxins. The clinical manifestations of these reactions are variable and unpredictable, including symptoms affecting the skin and the pulmonary, cardiac and gastrointestinal systems. The mechanism associated with their development is not yet fully understood. Diagnosis is based on patients' signs and symptoms and skin testing. The management of patients who suffer a hypersensitivity reaction to a chemotherapeutic agent varies with the severity of the reaction, the need to continue treatment, and the availability of alternative therapies. CONCLUSIONS: Due to a progressive increase in the use of chemotherapeutic agents an increased incidence of hypersensitivity reactions is to be expected. Desensitisation protocols are a noteworthy alternative that make it possible to re-initiate patients' therapy with the causative agent of the hypersensitivity reaction. Their use should be assessed individually, weighing risks and benefits.


Subject(s)
Antineoplastic Agents/adverse effects , Drug Hypersensitivity/etiology , Anaphylaxis/epidemiology , Anaphylaxis/etiology , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Antineoplastic Agents/immunology , Asparaginase/adverse effects , Asparaginase/immunology , Desensitization, Immunologic , Drug Eruptions/epidemiology , Drug Eruptions/etiology , Drug Hypersensitivity/epidemiology , Drug Hypersensitivity/immunology , Etoposide/adverse effects , Etoposide/immunology , Gastrointestinal Diseases/chemically induced , Gastrointestinal Diseases/epidemiology , Heart Diseases/chemically induced , Heart Diseases/epidemiology , Humans , Incidence , Organoplatinum Compounds/adverse effects , Organoplatinum Compounds/immunology , Recurrence , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/epidemiology , Risk , Taxoids/adverse effects , Taxoids/immunology
17.
Cancer ; 85(8): 1859-65, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10223582

ABSTRACT

BACKGROUND: Infants diagnosed with acute lymphoblastic leukemia (ALL) are considered the patient subgroup at the highest risk for central nervous system (CNS) disease, both at presentation and as an isolated extramedullary relapse. In addition, they are highly vulnerable to adverse developmental sequelae from CNS-directed therapy. METHODS: Thirty patients younger than 12 months at diagnosis (12 males, 18 females) in first hematologic remission were evaluated after completion of ALL therapy (mean age = 62.1 months; standard deviation = 17.2 months; range = 38-102 months). CNS-directed treatment included very high dose infusions of methotrexate (MTX) and intrathecal cytarabine and MTX. Three patients had meningeal leukemia that required additional therapy. Children were administered the McCarthy Scales of Children's Abilities, and parents completed a sociodemographic questionnaire to obtain information about occupation and education. RESULTS: Mean scores on all 6 cognitive and motor indices of the McCarthy Scales were in the average range (Verbal = 52.0; Perceptual = 53.6; Quantitative = 49.6; General Cognitive Index [GCI] = 102.1; Memory = 49.2; Motor = 51.0). Score distributions for each neurodevelopmental index were comparable to age-based population standards. One patient obtained a GCI that exceeded 2 standard deviations above the mean; none scored more than 2 standard deviations below. There was no report of developmental disabilities or neurologic disorders for any of the patients. Risk factors, including age at diagnosis, gender, additional CNS-directed treatment, and family socioeconomic status, were not associated with developmental outcome. CONCLUSIONS: Test findings indicated a generally positive neurodevelopmental outcome for ALL patients diagnosed in infancy who were treated with very high dose MTX as CNS-directed therapy. Combined with the reduction in the isolated CNS relapse rate achieved by the Children's Cancer Group (CCG) clinical trial CCG-107, the results of this study represent a substantial improvement in neurodevelopmental outcome for very young patients compared with infants treated for ALL in the past.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Damage, Chronic/etiology , Developmental Disabilities/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Survivors , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asparaginase/administration & dosage , Asparaginase/adverse effects , Brain Damage, Chronic/epidemiology , Cognition Disorders/epidemiology , Cognition Disorders/etiology , Combined Modality Therapy , Cranial Irradiation , Cytarabine/administration & dosage , Cytarabine/adverse effects , Daunorubicin/administration & dosage , Daunorubicin/adverse effects , Developmental Disabilities/epidemiology , Female , Follow-Up Studies , Humans , Infant , Injections, Spinal , Leucovorin/therapeutic use , Leukemic Infiltration/prevention & control , Male , Mercaptopurine/administration & dosage , Mercaptopurine/adverse effects , Methotrexate/administration & dosage , Methotrexate/adverse effects , Movement Disorders/epidemiology , Movement Disorders/etiology , Neuropsychological Tests , Precursor Cell Lymphoblastic Leukemia-Lymphoma/radiotherapy , Prednisone/administration & dosage , Prednisone/adverse effects , Psychomotor Performance , Remission Induction , Risk , Socioeconomic Factors , Vincristine/administration & dosage , Vincristine/adverse effects
18.
Leuk Res ; 21(9): 811-5, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9393595

ABSTRACT

Risk-directed chemotherapeutic regimens in recent use have improved the prognosis of children with acute lymphocytic leukemia (ALL). However, many patients relapse during or shortly after cessation of the initial continuation chemotherapy. Since achievement of a second complete remission (CR) is the initial step in successful retreatment effort, it is important to develop salvage protocols for children with relapsed or refractory ALL. In the present study, we developed a new salvage protocol (MLL-93) and applied the concept of dual chemical modulation of cytarabine, hydroxyurea, and etoposide with the alternative administration of high doses of myeloid- and lymphoid-directed agents. We also planned to perform allogeneic bone marrow transplantation (BMT) following a CR if patients had HLA-identical donor(s). The six patients treated with the MLL-93 protocol achieved a second CR. One patients in CR died of interstitial pneumonia after an unrelated allogeneic BMT. The other five patients have been in CR for 12-41 months. We suggest that the concepts of alternative administration of lymphoid- and myeloid-directed drugs and biochemical modulation are useful in the treatment of children with relapsed or refractory ALL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Salvage Therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Asparaginase/administration & dosage , Asparaginase/adverse effects , Asparaginase/pharmacology , Bone Marrow Transplantation , Child , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Cyclophosphamide/pharmacology , Cytarabine/administration & dosage , Cytarabine/adverse effects , Cytarabine/pharmacology , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Dexamethasone/pharmacology , Drug Administration Schedule , Drug Evaluation , Etoposide/administration & dosage , Etoposide/adverse effects , Etoposide/pharmacology , Female , Fever/etiology , Gastrointestinal Diseases/chemically induced , Humans , Hydrocortisone/administration & dosage , Hydrocortisone/adverse effects , Hydrocortisone/pharmacology , Hydroxyurea/administration & dosage , Hydroxyurea/adverse effects , Hydroxyurea/pharmacology , Leucovorin/administration & dosage , Leucovorin/adverse effects , Leucovorin/pharmacology , Male , Methotrexate/administration & dosage , Methotrexate/adverse effects , Methotrexate/pharmacology , Mitoxantrone/administration & dosage , Mitoxantrone/adverse effects , Mitoxantrone/pharmacology , Pilot Projects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Recurrence , Remission Induction , Treatment Outcome
19.
Cancer ; 80(12): 2285-95, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9404706

ABSTRACT

BACKGROUND: Infants with acute lymphoblastic leukemia (ALL) have a very poor prognosis. Since 1985, we have intensified therapy for infants with ALL by including a month of high dose multiagent chemotherapy after remission induction. METHODS: Between 1985 and 1995, we treated 23 infants (age < 12 months). We compared the presenting characteristics and outcomes of these infants with the 11 infants treated on our protocols between 1973 and 1985, an era prior to the intensification of therapy. Available bone marrow samples from infants treated since 1985 were analyzed for the presence of MLL gene rearrangements by Southern blot analyses and for TEL-AML1 gene fusion by reverse transcriptase-polymerase chain reaction. RESULTS: With a median follow-up of 5.6 years, the 50-month event free survival (EFS) (+/- standard error) for the 23 infants was 54 +/- 11%, a significant improvement (P = 0.001) compared with the outcome for the 11 infants treated on our protocols prior to 1985 (EFS = 9 +/- 9%). Of the seven infants found to have a rearranged MLL gene, three (43%) remained in first complete remission. None of the nine infant bone marrow specimens tested had evidence of TEL-AML1 gene fusion. The intensified therapy was complicated by a high incidence of infections, including septicemia in 52% of patients and Pneumocystis carinii pneumonitis in 22% of patients. Late effects identified in the 13 long term survivors (median age, 6 years) included developmental delay and learning disabilities of varying severity (82% of evaluable patients), asymptomatic cataracts (67%), asymptomatic echocardiographic abnormalities (30%), obesity (27%), and short stature (18%). CONCLUSIONS: Intensification of therapy significantly improved the EFS of infants with ALL compared with previous, less intensive regimens and with the experience of other investigators. Future treatment for infants should attempt to improve efficacy while minimizing toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Oncogene Proteins, Fusion , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogenes , Transcription Factors , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Asparaginase/administration & dosage , Asparaginase/adverse effects , Core Binding Factor Alpha 2 Subunit , Cytarabine/administration & dosage , Cytarabine/adverse effects , DNA, Neoplasm/analysis , DNA-Binding Proteins/genetics , Disease-Free Survival , Female , Histone-Lysine N-Methyltransferase , Humans , Infant , Infant, Newborn , Male , Mercaptopurine/administration & dosage , Mercaptopurine/adverse effects , Methotrexate/administration & dosage , Methotrexate/adverse effects , Myeloid-Lymphoid Leukemia Protein , Neoplasm Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Remission Induction , Survival Rate , Treatment Outcome , Vincristine/administration & dosage , Vincristine/adverse effects
20.
Blood Coagul Fibrinolysis ; 5(1): 23-8, 1994 Feb.
Article in English | MEDLINE | ID: mdl-7514043

ABSTRACT

Thrombotic events have been reported in acute lymphoblastic leukaemia patients, especially during or after L-asparaginase administration. A so-called L-asparaginase associated coagulopathy has been well recognized, being characterized by a hypercoagulable state (decrease of antithrombin III, plasminogen, protein C, protein S and increase of prothrombin fragment F1 + 2, thrombin-antithrombin complexes and fibrinopeptide A). The aim of this study was to determine whether the supplementation of antithrombin III (AT-III) concentrates could improve the L-asparaginase associated coagulopathy, thereby blocking the activation of the haemostatic system. In 25 adult patients with acute lymphoblastic leukaemia (M 19, F6, mean age 34 years) antithrombin III (AT-III) concentrates were administered at daily doses of 50 U/kg for 10 consecutive days from the beginning of L-asparaginase therapy (6,000 U/m2/day s.c. for 7 days), given according to the GIMEMA ALL 0288 trial. A marked increase of antithrombin III was recorded on days IV-VIII-XI (P < 0.001). No changes in protein C, protein S, plasminogen, alpha 2-antiplasmin, factor VII and platelet count were observed and there was no increase in markers of hypercoagulability. There was no evidence of disseminated intravascular coagulation. In conclusion, AT-III concentrate supplementation during L-asparaginase therapy, by the achievement of high levels of antithrombin III, is associated with a lack of activation of the haemostatic system and appears to overcome the complex coagulopathy associated with L-asparaginase.


Subject(s)
Antithrombin III/therapeutic use , Asparaginase/adverse effects , Blood Coagulation Disorders/drug therapy , Escherichia coli/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Antithrombin III/administration & dosage , Asparaginase/therapeutic use , Blood Coagulation Disorders/etiology , Cyclophosphamide/therapeutic use , Factor VII/metabolism , Female , Humans , Male , Middle Aged , Plasminogen/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Protein C/metabolism , Protein S/metabolism , alpha-2-Antiplasmin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL