Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Radiat Oncol ; 19(1): 34, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38475815

RESUMEN

BACKGROUND: FLASH therapy is a treatment technique in which radiation is delivered at ultra-high dose rates (≥ 40 Gy/s). The first-in-human FAST-01 clinical trial demonstrated the clinical feasibility of proton FLASH in the treatment of extremity bone metastases. The objectives of this investigation are to assess the toxicities of treatment and pain relief in study participants with painful thoracic bone metastases treated with FLASH radiotherapy, as well as workflow metrics in a clinical setting. METHODS: This single-arm clinical trial is being conducted under an FDA investigational device exemption (IDE) approved for 10 patients with 1-3 painful bone metastases in the thorax, excluding bone metastases in the spine. Treatment will be 8 Gy in a single fraction administered at ≥ 40 Gy/s on a FLASH-enabled proton therapy system delivering a single transmission proton beam. Primary study endpoints are efficacy (pain relief) and safety. Patient questionnaires evaluating pain flare at the treatment site will be completed for 10 consecutive days post-RT. Pain response and adverse events (AEs) will be evaluated on the day of treatment and on day 7, day 15, months 1, 2, 3, 6, 9, and 12, and every 6 months thereafter. The outcomes for clinical workflow feasibility are the occurrence of any device issues as well as time on the treatment table. DISCUSSION: This prospective clinical trial will provide clinical data for evaluating the efficacy and safety of proton FLASH for palliation of bony metastases in the thorax. Positive findings will support the further exploration of FLASH radiation for other clinical indications including patient populations treated with curative intent. REGISTRATION: ClinicalTrials.gov NCT05524064.


Asunto(s)
Neoplasias Óseas , Protones , Humanos , Neoplasias Óseas/radioterapia , Dolor , Estudios Prospectivos , Tórax
2.
Clin Oncol (R Coll Radiol) ; 33(11): 713-722, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34551871

RESUMEN

FLASH radiotherapy (FLASH-RT) delivers radiation treatment at an ultra-high dose rate that is several orders of magnitude higher than current clinical practice. In multiple preclinical studies, FLASH-RT has shown consistent normal tissue sparing effects while preserving equivalent antitumour activity in comparison with conventional dose rate radiation treatment. This is known as the 'FLASH effect'. Given the recent research interest in combining hypofractionated radiotherapy with immunotherapy to try to improve clinical outcomes, there is an intriguing clinical question as to whether FLASH irradiation may be a rational partner to combine with immune modulating drugs? To better predict the synergistic effect of both modalities, here we review the biological mechanisms of how FLASH differentially impacts the immune landscape, including circulating immune cells, tumour microenvironment and the inflammatory response. In order to make recommendations for future research, we summarise all published studies that investigated the immune modulatory effects of FLASH-RT and further explore the scientific reasons for combining FLASH with immunotherapy for potential clinical applications.


Asunto(s)
Neoplasias , Oncología por Radiación , Protocolos Clínicos , Humanos , Inmunoterapia , Neoplasias/radioterapia , Radioterapia , Dosificación Radioterapéutica
3.
Leukemia ; 22(2): 265-72, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18033323

RESUMEN

Polymorphisms of DNA repair genes RAD51 and XRCC3 increase susceptibility to acute myeloid leukemia (AML) in adults, an effect enhanced by deletion of the glutathione-S-transferase M1 (GSTM1) gene. In this study, we genotyped 452 children with de novo AML treated on CCG protocols 2941 and 2961 and compared genotype frequencies with those of normal blood donors, and analyzed the impact of genotype on outcome of therapy. XRCC3 Thr241Met, RAD51 G135C and GSTM1 genotypes did not increase susceptibility to AML when assessed singly. In contrast, when XRCC3 and RAD51 genotypes were examined together a significant increase in susceptibility to AML was seen in children with variant alleles. Analysis of outcome of therapy showed that patients heterozygous for the XRCC3 Thr241Met allele had improved post-induction disease-free survival compared to children homozygous for the major or minor allele, each of whom had similar outcomes. Improved survival was due to reduced relapse in the heterozygous children, and this effect was most marked in children randomized to therapy likely to generate DNA double-strand breaks (etoposide, daunomycin), compared with anti-metabolite (fludarabine, cytarabine) based therapy. In contrast, RAD51 G135C and the GSTM1 deletion polymorphism did not influence outcome of AML therapy in our study population.


Asunto(s)
Antineoplásicos/efectos adversos , Reparación del ADN/genética , Leucemia Mieloide Aguda/genética , Polimorfismo Genético/genética , Antineoplásicos/uso terapéutico , Proteínas de Unión al ADN/genética , Supervivencia sin Enfermedad , Frecuencia de los Genes , Genotipo , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/terapia , Pronóstico , Recombinasa Rad51/genética , Recurrencia , Resultado del Tratamiento
4.
Bone Marrow Transplant ; 37(2): 175-81, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16273111

RESUMEN

The prognosis for many pediatric and young adult patients with solid tumors that have metastasized at the time of diagnosis or have relapsed after therapy remains very poor. The steep dose-response curve of many of these tumors to alkylating agents makes myeloablative chemotherapy followed by autologous stem cell transplantation (ASCT) an attractive potential therapy. The role of ASCT for these high-risk patients is yet to be conclusively determined. We have transplanted 36 patients on two consecutive protocols with a variety of histological diagnoses. Overall survival (OS) was 63% (95% CI: 47-79%) at 1 year and 33% (95% CI: 16-50%) at 3 years. Patients with a diagnosis of Ewing's sarcoma (ES) or desmoplastic small round cell tumor (DSRCT) had significantly better survival than those with other diagnoses with estimated 3-year OS of 54% (95% CI: 29-79%) for this group of patients (P = 0.03). There were two transplant-related deaths both attributable to hepatic veno-occlusive disease. Median follow-up among survivors is 3.5 years (range: 0.6-7.9 years). These data justify continued investigation of ASCT as a consolidation therapy in patients with metastatic or relapsed ES and DSRCT.


Asunto(s)
Neoplasias Óseas/mortalidad , Neoplasias Óseas/terapia , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/terapia , Sarcoma de Ewing/mortalidad , Sarcoma de Ewing/terapia , Adolescente , Adulto , Neoplasias Óseas/complicaciones , Neoplasias Óseas/patología , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Fibroma Desmoplásico/complicaciones , Fibroma Desmoplásico/mortalidad , Fibroma Desmoplásico/patología , Fibroma Desmoplásico/terapia , Estudios de Seguimiento , Enfermedad Veno-Oclusiva Hepática/etiología , Enfermedad Veno-Oclusiva Hepática/mortalidad , Humanos , Masculino , Recurrencia Local de Neoplasia/complicaciones , Recurrencia Local de Neoplasia/patología , Factores de Riesgo , Sarcoma de Ewing/complicaciones , Sarcoma de Ewing/patología , Trasplante de Células Madre/métodos , Trasplante de Células Madre/mortalidad , Tasa de Supervivencia , Trasplante Autólogo
5.
Leukemia ; 18(4): 685-92, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14990973

RESUMEN

Activating mutations in the RAS oncogenes are among the most common genetic alterations in human cancers, including patients with acute lymphoblastic leukemia (ALL). We sought to define the frequency and spectrum, and possible prognostic importance, of N- and K-RAS mutations in children with ALL treated with contemporary therapy. Leukemic blast DNA from 870 children was analyzed for the presence of activating mutations in the N- or K-RAS oncogenes using a sensitive mutation detection algorithm. RAS mutations were present in the blasts of 131 (15.1%) pediatric ALL patients. The spectrum of mutations included 81 (9.3%) mutations of codons 12/13 of N-RAS, 12 (1.4%) mutations of codon 61 of N-RAS, 39 (4.5%) mutations of codons 12/13 of K-RAS, and 2 (0.2%) mutations of codon 61 of K-RAS. The presence of N- or K-RAS mutations was not associated with white blood cell count at diagnosis, sex, race, extramedullary testicular involvement, central nervous system disease, or NCI/CTEP ALL Risk Group. Patients with an exon 1 K-RAS mutation (codons 12/13) were significantly younger at diagnosis (P=0.001) and less frequently B-lineage phenotype (P=0.01). RAS mutation status did not predict overall survival, event-free survival and disease-free survival. While N- and K-RAS mutations can be identified in 15% of children with newly diagnosed ALL, they do not represent a significant risk factor for outcome using contemporary chemotherapy regimens.


Asunto(s)
Genes ras/genética , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Preescolar , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Epidemiología Molecular , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/epidemiología , Pronóstico , Factores de Riesgo , Análisis de Supervivencia , Resultado del Tratamiento
6.
Ann Oncol ; 13 Suppl 1: 34-9, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12078900

RESUMEN

Survivors of childhood Hodgkin's lymphoma (HL) have an increased risk of developing treatment-related complications, especially second malignant neoplasms, as a result of treatment regimens incorporating chemotherapy and radiation therapy. Second cancers include leukemias that generally occur in the first two decades after therapy, and adult-type solid tumors that generally exhibit continued increasing incidence throughout subsequent follow-up. Identified clinical risk factors for second cancers include age at the time of treatment and intensity and type of therapy, with particularly strong associations between the use of radiotherapy and subsequent breast cancer, and alkylator chemotherapy dose-intensity and risk of secondary leukemia. However, second cancers affect a minority of patients, and there is probably great variability in individual susceptibility for this complication. Common genetic polymorphisms in drug-metabolizing enzymes that result in impaired detoxification of chemotherapy or inefficient repair of drug- or radiation-induced genetic damage may lead to increased risk of a second cancer. Studies of the potential role of polymorphisms in the genes encoding the glutathione S-transferases, cytochrome P450 3A4, NAD(P)H:quinone oxidoreductase and myeloperoxidase in the etiology of treatment-related complications are reviewed. Biological markers of drug- and radiation-induced genetic damage may also identify patients at higher risk of immediate and delayed side effects of therapy. The Children's Oncology Group (COG) is examining the roles of polymorphisms in drug metabolizing enzymes and biological markers of genotoxicity in predicting the treatment-related outcomes of patients with HL. These investigations may ultimately allow the use of pharmacogenetically guided therapy to improve the outcome of HL therapy and reduce the risk of therapy-related complications, especially secondary malignancies.


Asunto(s)
Antineoplásicos/uso terapéutico , Sistema Enzimático del Citocromo P-450/genética , Marcadores Genéticos , Glutatión Transferasa/genética , Enfermedad de Hodgkin/enzimología , NAD(P)H Deshidrogenasa (Quinona)/genética , Neoplasias Primarias Secundarias/etiología , Peroxidasa/genética , Polimorfismo Genético , Citocromo P-450 CYP3A , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/terapia , Humanos
7.
Hematol Oncol Clin North Am ; 15(4): 677-701, viii-ix, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11676279

RESUMEN

Approximately half of children with acute myeloid leukemia (AML) can be cured with contemporary chemotherapy regimens; however, various forms of drug resistance pose considerable obstacles for curing the remaining patients. Recent advances in immunology, cytogenetics, and cellular and molecular biology have provided new insights into fundamental biological differences between leukemic myeloid blasts and their normal counterparts. This article focuses on new technologies involving: (1) antibody- or growth factor-mediated targeting of antigens or growth factor receptors found on AML blasts and restricted sub-groups of normal cells, (2) pharmacologic targeting of the pathologic t(15;17) translocation of acute promyelocytic leukemia with all-trans retinoic acid, (3) pharmacologic and immunologic targeting of mutant RAS oncogenes and related aberrant signaling in AML blasts, and (4) targeting of pathological signaling of the Bcr-Abl oncoprotein and c-kit tyrosine kinase in myeloid leukemias. These advances herald an exciting new era of AML-specific therapies.


Asunto(s)
Aminoglicósidos , Leucemia Mieloide/tratamiento farmacológico , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/metabolismo , Enfermedad Aguda , Adulto , Animales , Antibacterianos/uso terapéutico , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Antígenos de Neoplasias/efectos de los fármacos , Antígenos de Neoplasias/inmunología , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Niño , Preescolar , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Terapia Combinada , Diseño de Fármacos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Predicción , Gemtuzumab , Humanos , Inmunoconjugados/uso terapéutico , Inmunotoxinas/uso terapéutico , Lactante , Leucemia Mieloide/mortalidad , Leucemia Mieloide/radioterapia , Ratones , Ratones SCID , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/genética , Riesgo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Med Pediatr Oncol ; 36(5): 541-8, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11340609

RESUMEN

Treatment-related leukemias are one of the most devastating late complications of cancer therapy. Patients with rare cancer predisposition syndromes including neurofibromatosis type 1 and inherited p53 mutations are at an increased risk for this complication. Other patients may have increased susceptibility because they possess common genetic polymorphisms in drug-metabolizing enzymes that result in impaired detoxification of chemotherapy or inefficient repair of drug-induced genetic damage. We review studies that have identified a potential role for polymorphisms in the genes encoding the glutathione-S-transferases (GSTs), NAD(P) H: quinone oxidoreductase, myeloperoxidase, N-acetyltransferase (NATs), cytochrome P450 (CYP) 1A1 and 3A4, methylenetetrahydrofolate reductase (MTHFR), cystathionine-beta-synthase (CBS), and others in the etiology of primary or secondary acute leukemias, and therapy-related complications. The identification of high risk polymorphisms and use of pharmacogenetically-guided therapies holds promise to improve the outcome of cancer therapy and reduce the risk of treatment-related leukemias.


Asunto(s)
Predisposición Genética a la Enfermedad , Leucemia/diagnóstico , Leucemia/genética , Glutatión Transferasa/genética , Humanos , Leucemia/enzimología , Polimorfismo Genético
9.
J Clin Oncol ; 19(5): 1279-87, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11230469

RESUMEN

PURPOSE: Glutathione S-transferase theta (GSTT1) and mu (GSTM1) genes are polymorphic, the genes being absent in approximately 15% and 50% of the population, respectively. Because glutathione S-transferases may be involved in the metabolism of chemotherapy drugs, we hypothesized that presence or absence of the genes may influence the outcome of treatment for childhood acute myeloid leukemia (AML). PATIENTS AND METHODS: We genotyped GSTT1 and GSTM1 in 306 children with AML receiving chemotherapy on Children's Cancer Group therapeutic studies. Outcomes were compared in those with and without GSTT1 and GSTM1 genes. RESULTS: Patients with the GSTT1-negative genotype had reduced survival compared with those with at least one GSTT1 allele (GSTT1 positive) (52% v 40% at 5 years; log-rank P =.05). A multivariate model of survival adjusted for age group, sex, WBC count, chloroma, CNS involvement, and French-American-British group confirmed the increased risk of death in the GSTT1-null cases (relative risk, AQ 1.6; P =.02). The frequency of death in remission was increased in GSTT1-negative cases compared with GSTT1-positive cases (24% v 12%, log-rank P =.05). The frequency of relapse from end of induction was similar in GSTT1-negative and GSTT1-positive cases (38% v 35%, log-rank P =.5). CONCLUSION: Children who lacked GSTT1 had greater toxicity and reduced survival after chemotherapy for AML compared with children with at least one GSTT1 allele. If confirmed in further studies, GSTT1 genotype might be useful in selecting appropriate chemotherapy regimens for children with AML.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Glutatión Transferasa/genética , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/genética , Polimorfismo Genético , Niño , Preescolar , ADN de Neoplasias/genética , Femenino , Genotipo , Humanos , Masculino , Reacción en Cadena de la Polimerasa , Pronóstico , Recurrencia , Medición de Riesgo , Análisis de Supervivencia
10.
Methods Mol Biol ; 166: 31-53, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11217375

Asunto(s)
Toxina Diftérica/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Inmunotoxinas/uso terapéutico , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/uso terapéutico , Adulto , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Niño , Preescolar , Clonación Molecular/métodos , Pruebas Inmunológicas de Citotoxicidad , Toxina Diftérica/genética , Toxina Diftérica/aislamiento & purificación , Toxina Diftérica/farmacología , Diseño de Fármacos , Resistencia a Antineoplásicos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/aislamiento & purificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Humanos , Inmunotoxinas/genética , Inmunotoxinas/aislamiento & purificación , Inmunotoxinas/farmacología , Lactante , Interleucina-2/uso terapéutico , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Proteínas de Neoplasias/fisiología , Poli(ADP-Ribosa) Polimerasas/análisis , Subunidades de Proteína , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas/efectos de los fármacos
11.
Cancer Epidemiol Biomarkers Prev ; 9(6): 563-6, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10868689

RESUMEN

GSTM1 and GSTT1 are polymorphic genes. Absence of enzyme activity is due to homozygous inherited deletion of the gene, reducing detoxification of carcinogens such as epoxides and alkylating agents and potentially increasing cancer risk. We hypothesized that GST null genotype would increase risk of acute myeloid leukemia and myelodysplasia (AML/MDS) in children. DNA was extracted from bone marrow slides of 292 AML/MDS patients. PCR amplification was used to assign GSTM1 and GSTT1 genotypes for cases and controls. Given that the frequency of the null genotype varies by ethnicity and that the majority of the cases were Caucasian, analyses were restricted to 232 white (non-Hispanic) cases and 153 Caucasian non cancer controls. The frequency of GSTM1 null was significantly increased in AML/MDS cases compared with controls [64 versus 47%; odds ratio (OR), 2.0 [95% confidence interval (CI), 1.3-3.1]; P = 0.001], whereas the frequency of GSTT1 null genotype in AML/MDS cases was not statistically different from controls. AML comprises biologically distinct subtypes, and a test for homogeneity revealed a statistically significant difference among subtypes (P = 0.04; df, 8) for GSTM1 only. In particular, there was an increased frequency of GSTM1 null genotypes in French-American-British groups M3 [82%; n = 22; OR, 5.1 (95% CI, 1.6-21.3)] and M4 [72%; n = 53; OR, 2.9 (95% CI, 1.4-6.0)]. We conclude that the GSTM1 null genotype is a significant risk factor for childhood AML, particularly French-American-British groups M3 and M4. This may indicate an important role for exogenous carcinogens in the etiology of childhood AML.


Asunto(s)
Glutatión Transferasa/genética , Leucemia Mieloide/enzimología , Síndromes Mielodisplásicos/enzimología , Polimorfismo Genético , Niño , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Lactante , Leucemia Mieloide/genética , Síndromes Mielodisplásicos/genética , Reacción en Cadena de la Polimerasa , Factores de Riesgo
12.
Cancer ; 85(6): 1389-94, 1999 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10189147

RESUMEN

BACKGROUND: Insulin-like growth factor-2 (IGF2) and H19 are reciprocally imprinted genes on chromosome 11; IGF2 is expressed paternally and H19 is expressed maternally. Loss of imprinting (LOI) at both H19 and IGF2 has been reported in seven fully informative adult testicular germ cell tumors (GCTs) and may contribute to germ cell carcinogenesis. METHODS: Genomic DNA from 61 pediatric GCTs was amplified by polymerase chain reaction (PCR) and screened for heterozygosity at both IGF2 and H19 using either ApaI or RsaI, respectively. If heterozygous, polyadenylated RNA was isolated and reversed-transcribed into cDNA. cDNA then was amplified by PCR and the products were digested with restriction enzymes to evaluate GCT expression of IGF2 and H19. RESULTS: Eleven pediatric GCTs were fully informative for H19 and IGF2, including 5 ovarian GCTs, 2 testicular GCTs, and 4 extragonadal GCTs. Consistent with prior studies, both testicular GCTs showed LOI at both H19 and IGF2. In contrast, three of the five ovarian GCTs had LOI at both IGF2 and H19; one had LOI at IGF2 only, and one retained imprinting at both loci. Only one of the four extragonadal GCTs had LOI at IGF2 whereas three of the four had LOI at H19. CONCLUSIONS: These data suggest that LOI at H19 and IGF2 also may be common in pediatric testicular GCTs. However, ovarian and extragonadal pediatric GCTs showed variable patterns of LOI that may indicate differences in the timing of carcinogenesis in germ cells at these sites.


Asunto(s)
Impresión Genómica , Germinoma/genética , Proteínas Musculares/genética , ARN no Traducido , Adolescente , Adulto , Niño , Preescolar , Femenino , Heterocigoto , Humanos , Lactante , Recién Nacido , Masculino , Proteínas Musculares/análisis , Neoplasias Ováricas/química , Neoplasias Ováricas/genética , Reacción en Cadena de la Polimerasa , Polimorfismo Genético , ARN Largo no Codificante , Neoplasias Testiculares/química , Neoplasias Testiculares/genética
13.
Cancer Epidemiol Biomarkers Prev ; 7(12): 1127-31, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9865432

RESUMEN

Archival slides are a potentially useful source of DNA for mutation analyses in large population-based studies. However, it is unknown whether specimen age or histological stains alter the accuracy of Taq polymerase or induce secondary mutations in sample DNA. To address this question, we evaluated five methods for extraction of genomic DNA from archival bone marrow slides of 17 leukemia patients and analyzed exons 1 and 2 of the N- and K-ras genes for the presence of mutations. Of the five methods, optimal DNA purification was achieved by boiling and phenol:chloroform extraction. N-and K-ras exons 1 and 2 were independently amplified using 35 cycles of PCR, and 6-12 clones for each exon were isolated and individually sequenced for each patient. Mutations were confirmed by repeat extraction, cloning, and sequencing. Sixteen of 17 patient samples were successfully amplified (94%), including slides up to 29 years old. Twelve slides had been stained with Wright-Giemsa, I stained with toluidine blue, and 4 were unstained. A total of 16 single-base mutations were identified of 33,840 nucleotides sequenced. No insertions or deletions were identified. Six of 16 single-base mutations were previously described activating mutations in codon 13 of N-ras exon 1. The 10 other mutations were in other regions of the N- and K-ras genes and were not reproduced after repeat extraction, cloning, and sequencing. The frequency of these other alterations was I of 3384 bp. This value is comparable with the inherent error frequency for Taq polymerase. Our findings suggest that high fidelity DNA amplification can be achieved using archival hematological slides as old as 29 years and can be reliably used in genetic analyses.


Asunto(s)
Médula Ósea , ADN/análisis , Leucemia/genética , Reacción en Cadena de la Polimerasa , Archivos , Niño , Marcadores Genéticos , Humanos , Proyectos Piloto , Reacción en Cadena de la Polimerasa/métodos , Reproducibilidad de los Resultados , Factores de Tiempo , Conservación de Tejido
14.
J Pediatr ; 132(5): 863-5, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9602201

RESUMEN

Hemoglobin E (HbE), alpha-thalassemia, and beta-thalassemia are common among Southeast Asians and often occur in compound heterozygous states that complicate neonatal screening. We describe a kindred with alpha-thalassemia-1, HbE, and beta zero-thalassemia. The proband had HbE/beta zero-thalassemia, with severe anemia and failure to thrive. His father also had HbE/beta zero-thalassemia but had coinherited alpha-thalassemia-1 and was free of symptoms.


Asunto(s)
Hemoglobina E/genética , Talasemia alfa/genética , Talasemia beta/genética , Adulto , Asia Sudoriental/etnología , Preescolar , Femenino , Asesoramiento Genético , Genotipo , Heterocigoto , Humanos , Recién Nacido , Masculino , Tamizaje Neonatal , Talasemia alfa/complicaciones , Talasemia alfa/etnología , Talasemia beta/complicaciones , Talasemia beta/etnología
15.
Leukemia ; 11 Suppl 4: S4-7, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9179272

RESUMEN

Despite the use of similar intensive chemotherapy regimens, adults with acute lymphoblastic leukemia (ALL) exhibit a strikingly inferior outcome when compared to children. Mirroring this difference in prognosis, childhood and adult ALL exhibit distinctive age-related differences in potential etiologic factors and underlying biology. Childhood ALL mostly occurs in industrialized countries, with a unique peak in incidence between the ages of 2 and 8 years. It is associated with features conferring a favorable response to therapy, including early pre-B cell disease and a hyperdiploid karyotype or a cryptic t(12;21) translocation. The lymphoblasts of childhood ALL appear to arise in a developmental compartment that is "poised" for apoptotic death and are particularly sensitive to glucocorticoids, antimetabolites, and other cytotoxic agents. In contrast, adult ALL commonly possesses the poor-prognosis Philadelphia chromosome and is often drug-resistant. A far higher proportion of adults with ALL present with high initial white blood cell count and high-risk immunophenotypes, and exhibit a slow induction of remission with greater therapy-related toxicity. Improved supportive care measures with current intensive therapies and the development of novel leukemia-targeted biotherapies will be required to improve the cure rates of adults with ALL.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Adulto , Factores de Edad , Niño , Preescolar , Cromosomas Humanos Par 12 , Cromosomas Humanos Par 21 , Humanos , Incidencia , Cariotipificación , Leucemia-Linfoma Linfoblástico de Células Precursoras/epidemiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Pronóstico , Translocación Genética
16.
Leuk Lymphoma ; 25(3-4): 247-56, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9168435

RESUMEN

Contemporary therapies for acute myeloid leukemia (AML) commonly fail to cure patients because of the emergence of drug resistance. Drug resistance in AML is multifactorial but can be associated with the overexpression of transmembrane transporter molecules, including P-glycoprotein (Pgp) or the multidrug resistance-associated protein (MRP), or associated with inactivation of the p53 tumor suppressor gene, as well as overexpression of the anti-apoptotic protein bcl-2. We are investigating if novel recombinant biotherapeutics can circumvent these resistance mechanisms to effectively treat refractory AML. To target the lethal action of diphtheria toxin (DT) to high affinity granulocyte-macrophage colony-stimulating factor (GMCSF) receptors on AML blasts, we have produced a recombinant chimeric fusion toxin, DTctGMCSF. Since DTctGMCSF enters and kills its target cells by unique mechanisms (GMCSF-receptor binding and protein synthesis inhibition) and is not similar in structure to Pgp or MRP substrates, we postulated that it would be an active agent against therapy-resistant AML. DTctGMCSF was selectively cytotoxic (IC50 1-10ng/ml) to GMCSF-receptor positive AML cells expressing the Pgp- or MRP-associated multi-drug resistant phenotypes, despite high level resistance to conventional chemotherapeutic agents. DTctGMCSF also efficiently killed AML cells deficient in p53 expression, as well as radiation-resistant AML cells and mixed lineage leukemia cells expressing high levels of bcl-2. In addition, DTctGMCSF killed > 99% of primary leukemic progenitor cells from therapy-refractory AML patients under conditions that we have previously found to not adversely affect the proliferative capacity or differentiation of pluripotent normal hematopoietic progenitor cells. DTctGMCSF may prove useful in treating myeloid leukemias that are otherwise resistant to a wide range of conventional therapies.


Asunto(s)
Toxina Diftérica/farmacología , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/radioterapia , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Antineoplásicos/farmacología , Toxina Diftérica/genética , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Leucemia Mieloide/patología , Tolerancia a Radiación , Proteínas Recombinantes de Fusión/genética , Células Madre/efectos de los fármacos , Células Tumorales Cultivadas
17.
Leuk Lymphoma ; 25(3-4): 257-70, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9168436

RESUMEN

Human granulocyte-macrophage colony stimulating factor (GMCSF) and its high affinity receptor function to regulate the proliferation and differentiation of myeloid lineage hematopoietic cells, and may participate in the pathogenesis of many malignant myeloid diseases. We have used genetic engineering based on the elucidated molecular structures of human granulocyte-macrophage colony-stimulating factor and diphtheria toxin (DT) to produce a recombinant fusion toxin, DTctGMCSF, that targets diphtheria toxin to high affinity GMCSF receptors expressed on the surface of blast cells from a large fraction of patients with acute myeloid leukemia (AML). DTctGMCSF was specifically immunoreactive with antidiphtheria toxin and anti-GMCSF antiseras, and exhibited the characteristic catalytic activity of diphtheria toxin, catalyzing the in vitro ADP-ribosylation of purified elongation factor 2. The cytotoxic effects of DTctGMCSF were examined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-tetrazolium (MTT) bromide assay of cell viability and in vivo assays of protein synthesis inhibition. DTctGMCSF were specifically cytotoxic to human leukemia cell lines bearing high affinity receptors for human GMCSF with IC50 of 10(-9) to 10(-11) M. It was not toxic to mammalian hematopoietic cell lines lacking human GMCSF (hGMCSF) receptors. In receptor positive cells, cytotoxicity can be specifically blocked by a large excess of hGMCSF, confirming that its cytotoxicity is mediated through the hGMCSF receptor. THough DTctGMCSF inhibited granulocyte-macrophage colony formation by committed myeloid progenitor cells (CFU-GM), it did not significantly affect erythroid burst formation by committed erythroid progenitor cells (BFU-E), or mixed granulocyte-erythroid-macrophage-megakaryocyte colony formation by pluripotent multilineage progenitor cells (CFU-GEMM). DTctGMCSF holds promise for the treatment of myeloid lineage malignancies, and is a useful reagent to study hematopoiesis.


Asunto(s)
Toxina Diftérica/farmacología , Leucemia/tratamiento farmacológico , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Animales , Antineoplásicos/farmacología , Médula Ósea/efectos de los fármacos , Células de la Médula Ósea , División Celular/efectos de los fármacos , Toxina Diftérica/genética , Toxina Diftérica/inmunología , Ensayos de Selección de Medicamentos Antitumorales , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Sueros Inmunes , Leucemia/genética , Leucemia/patología , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/patología , Ratones , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteínas Recombinantes de Fusión/genética , Células Madre/efectos de los fármacos , Células Tumorales Cultivadas
18.
Clin Cancer Res ; 3(3): 347-55, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9815691

RESUMEN

Multiagent chemotherapy regimens fail to cure more than one-half of the patients with acute myeloid leukemia (AML) because of the emergence of dominant multidrug-resistant subclones of leukemia cells. We have developed a recombinant diphtheria toxin-human granulocyte macrophage colony-stimulating factor chimeric fusion protein (DTctGMCSF) that specifically targets GMCSF receptor-positive AML cells. This novel biotherapeutic agent induced rapid apoptotic cell death of chemotherapy-resistant AML cell lines and primary leukemic cells from treatment-refractory AML patients. Our results suggest that DTctGMCSF may be useful in the treatment of AML patients whose leukemia has recurred and developed resistance to contemporary chemotherapy programs.


Asunto(s)
Apoptosis/fisiología , Toxina Diftérica/toxicidad , Resistencia a Múltiples Medicamentos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/toxicidad , Inmunotoxinas/toxicidad , Tolerancia a Radiación , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/toxicidad , Enfermedad Aguda , Apoptosis/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Células HL-60 , Humanos , Leucemia Mieloide , Leucemia Mieloide Aguda , Células Tumorales Cultivadas
19.
Clin Cancer Res ; 3(12 Pt 1): 2217-27, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9815618

RESUMEN

Acute myeloid leukemia (AML) is the most common form of acute leukemia. Contemporary chemotherapy regimens fail to cure most patients with AML. We have genetically engineered a recombinant diphtheria toxin human granulocyte macrophage colony-stimulating factor (GMCSF) chimeric fusion protein (DTctGMCSF) that specifically targets the GMCSF receptor on fresh human AML cells and myeloid leukemia cell lines. At a nontoxic dose level, DTctGMCSF therapy was superior to the standard chemotherapeutic agents 1-beta-D-arabinofuranosylcytosine and Adriamycin, resulting in 60% long-term event-free survival of severe combined immunodeficient mice challenged with an otherwise invariably fatal cell dose of the human HL-60 myeloid leukemia. Notably, systemic exposure levels of DTctGMCSF, which were found to be therapeutic in the severe combined immunodeficient mouse xenograft model of human HL-60 myeloid leukemia, could be achieved in cynomolgus monkeys without any significant nonhematological toxicities. The recombinant DTctGMCSF fusion toxin might be useful in the treatment of AML patients whose leukemias have recurred and developed resistance to contemporary chemotherapy programs.


Asunto(s)
Toxina Diftérica/farmacocinética , Toxina Diftérica/uso terapéutico , Inmunotoxinas/farmacocinética , Inmunotoxinas/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Animales , Citarabina/uso terapéutico , Toxina Diftérica/toxicidad , Supervivencia sin Enfermedad , Doxorrubicina/uso terapéutico , Femenino , Células HL-60 , Humanos , Inmunotoxinas/toxicidad , Macaca fascicularis , Ratones , Ratones SCID , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Recombinantes de Fusión/toxicidad , Distribución Tisular , Trasplante Heterólogo
20.
Cancer Causes Control ; 7(5): 553-9, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8877054

RESUMEN

Several epidemiologic studies have demonstrated that high birthweight is associated with an increased risk of infant leukemia; however, the reason for this relationship is unclear. Biologic data demonstrate that birth weight is correlated positively with circulating levels of insulin-like growth factor-1 (IGF-1). IGF-1 is important in blood formation and regulation and has been shown to stimulate the growth of both myeloid and lymphoid cells in culture. Since infants who develop leukemia are likely to have had at least one transforming event occur in utero, we hypothesize that high levels of IGF-1 may both produce a larger baby and contribute to leukemogenesis.


Asunto(s)
Peso al Nacer , Factor I del Crecimiento Similar a la Insulina/fisiología , Leucemia/etiología , Células de la Médula Ósea , División Celular , Células Cultivadas , Hematopoyesis/fisiología , Humanos , Lactante , Recién Nacido , Factor I del Crecimiento Similar a la Insulina/análisis , Leucemia/epidemiología , Linfocitos/citología , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA