Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 39(6): 110805, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35545056

RESUMEN

Myelodysplastic syndrome (MDS) is a clonal disorder of hematopoietic stem cells (HSCs), characterized by ineffective hematopoiesis and frequent progression to leukemia. It has long remained unresolved how MDS cells, which are less proliferative, inhibit normal hematopoiesis and eventually dominate the bone marrow space. Despite several studies implicating mesenchymal stromal or stem cells (MSCs), a principal component of the HSC niche, in the inhibition of normal hematopoiesis, the molecular mechanisms underlying this process remain unclear. Here, we demonstrate that both human and mouse MDS cells perturb bone metabolism by suppressing the osteolineage differentiation of MSCs, which impairs the ability of MSCs to support normal HSCs. Enforced MSC differentiation rescues the suppressed normal hematopoiesis in both in vivo and in vitro MDS models. Intriguingly, the suppression effect is reversible and mediated by extracellular vesicles (EVs) derived from MDS cells. These findings shed light on the novel MDS EV-MSC axis in ineffective hematopoiesis.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Síndromes Mielodisplásicos , Animales , Vesículas Extracelulares/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Síndromes Mielodisplásicos/metabolismo
2.
iScience ; 24(6): 102651, 2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34151238

RESUMEN

A hallmark of acute myeloid leukemia (AML) is the inability of self-renewing malignant cells to mature into a non-dividing terminally differentiated state. This differentiation block has been linked to dysregulation of multiple cellular processes, including transcriptional, chromatin, and metabolic regulation. The transcription factor HOXA9 and the histone demethylase LSD1 are examples of such regulators that promote differentiation blockade in AML. To identify metabolic targets that interact with LSD1 inhibition to promote myeloid maturation, we screened a small molecule library to identify druggable substrates. We found that differentiation caused by LSD1 inhibition is enhanced by combined perturbation of purine nucleotide salvage and de novo lipogenesis pathways, and identified multiple lines of evidence to support the specificity of these pathways and suggest a potential basis of how perturbation of these pathways may interact synergistically to promote myeloid differentiation. In sum, these findings suggest potential drug combination strategies in the treatment of AML.

3.
Cancer Discov ; 11(6): 1542-1561, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33500244

RESUMEN

Patients with acute myeloid leukemia (AML) frequently relapse after chemotherapy, yet the mechanism by which AML reemerges is not fully understood. Herein, we show that primary AML cells enter a senescence-like phenotype following chemotherapy in vitro and in vivo. This is accompanied by induction of senescence/inflammatory and embryonic diapause transcriptional programs, with downregulation of MYC and leukemia stem cell genes. Single-cell RNA sequencing suggested depletion of leukemia stem cells in vitro and in vivo, and enrichment for subpopulations with distinct senescence-like cells. This senescence effect was transient and conferred superior colony-forming and engraftment potential. Entry into this senescence-like phenotype was dependent on ATR, and persistence of AML cells was severely impaired by ATR inhibitors. Altogether, we propose that AML relapse is facilitated by a senescence-like resilience phenotype that occurs regardless of their stem cell status. Upon recovery, these post-senescence AML cells give rise to relapsed AMLs with increased stem cell potential. SIGNIFICANCE: Despite entering complete remission after chemotherapy, relapse occurs in many patients with AML. Thus, there is an urgent need to understand the relapse mechanism in AML and the development of targeted treatments to improve outcome. Here, we identified a senescence-like resilience phenotype through which AML cells can survive and repopulate leukemia.This article is highlighted in the In This Issue feature, p. 1307.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Células Madre Neoplásicas/citología , Inducción de Remisión , Animales , Línea Celular Tumoral/citología , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Recurrencia Local de Neoplasia/patología , Fenotipo
4.
Blood ; 137(6): 812-825, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32911532

RESUMEN

B-cell lymphoma 6 (BCL6) is a transcription repressor and proto-oncogene that plays a crucial role in the innate and adaptive immune system and lymphoid neoplasms. However, its role in myeloid malignancies remains unclear. Here, we explored the role of BCL6 in acute myeloid leukemia (AML). BCL6 was expressed at variable and often high levels in AML cell lines and primary AML samples. AMLs with higher levels of BCL6 were generally sensitive to treatment with BCL6 inhibitors, with the exception of those with monocytic differentiation. Gene expression profiling of AML cells treated with a BCL6 inhibitor revealed induction of BCL6-repressed target genes and transcriptional programs linked to DNA damage checkpoints and downregulation of stem cell genes. Ex vivo treatment of primary AML cells with BCL6 inhibitors induced apoptosis and decreased colony-forming capacity, which correlated with the levels of BCL6 expression. Importantly, inhibition or knockdown of BCL6 in primary AML cells resulted in a significant reduction of leukemia-initiating capacity in mice, suggesting ablation of leukemia repopulating cell functionality. In contrast, BCL6 knockout or inhibition did not suppress the function of normal hematopoietic stem cells. Treatment with cytarabine further induced BCL6 expression, and the levels of BCL6 induction were correlated with resistance to cytarabine. Treatment of AML patient-derived xenografts with BCL6 inhibitor plus cytarabine suggested enhanced antileukemia activity with this combination. Hence, pharmacologic inhibition of BCL6 might provide a novel therapeutic strategy for ablation of leukemia-repopulating cells and increased responsiveness to chemotherapy.


Asunto(s)
Leucemia Mieloide Aguda/patología , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas c-bcl-6/fisiología , Animales , Antineoplásicos/farmacología , Apoptosis , Autorrenovación de las Células , Citarabina/uso terapéutico , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Madre Hematopoyéticas/citología , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Leucemia Mieloide Aguda/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/citología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-bcl-6/antagonistas & inhibidores , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , RNA-Seq , Quimera por Radiación , Tiazolidinedionas/farmacología , Tiazolidinedionas/uso terapéutico , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Biochem ; 159(1): 17-25, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26590301

RESUMEN

Recent progress in high-speed sequencing technology has revealed that tumors harbor novel mutations in a variety of genes including those for molecules involved in epigenetics and splicing, some of which were not categorized to previously thought malignancy-related genes. However, despite thorough identification of mutations in solid tumors and hematological malignancies, how these mutations induce cell transformation still remains elusive. In addition, each tumor usually contains multiple mutations or sometimes consists of multiple clones, which makes functional analysis difficult. Fifteen years ago, it was proposed that combination of two types of mutations induce acute leukemia; Class I mutations induce cell growth or inhibit apoptosis while class II mutations block differentiation, co-operating in inducing acute leukemia. This notion has been proven using a variety of mouse models, however most of recently found mutations are not typical class I/II mutations. Although some novel mutations have been found to functionally work as class I or II mutation in leukemogenesis, the classical class I/II theory seems to be too simple to explain the whole story. We here overview the molecular basis of hematological malignancies based on clinical and experimental results, and propose a new working hypothesis for leukemogenesis.


Asunto(s)
Carcinogénesis/genética , Neoplasias Hematológicas/genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Translocación Genética , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Epigénesis Genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Mutación , Proteínas de Fusión Oncogénica/genética , Fenotipo
6.
Pulm Circ ; 5(2): 382-97, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26064466

RESUMEN

Although there are many studies focusing on the molecular pathways underlying lung vascular morphogenesis, the extracellular matrix (ECM)-dependent regulation of mesenchymal cell differentiation in vascular smooth muscle development needs better understanding. In this study, we demonstrate that the paired related homeobox gene transcription factor Prx1 maintains the elastic ECM properties, which are essential for vascular smooth muscle precursor cell differentiation. We have found that Prx1(null) mouse lungs exhibit defective vascular smooth muscle development, downregulated elastic ECM expression, and compromised transforming growth factor (TGF)-ß localization and signaling. Further characterization of ECM properties using decellularized lung ECM scaffolds derived from Prx1 mice demonstrated that Prx1 is required to maintain lung ECM stiffness. The results of cell culture using stiffness-controlled 2-D and 3-D synthetic substrates confirmed that Prx1-dependent ECM stiffness is essential for promotion of smooth muscle precursor differentiation for effective TGF-ß stimulation. Supporting these results, both decellularized Prx1(null) lung ECM and Prx1(WT) (wild type) ECM scaffolds with blocked TGF-ß failed to support mesenchymal cell to 3-D smooth muscle cell differentiation. These results suggest a novel ECM-dependent regulatory pathway of lung vascular development wherein Prx1 regulates lung vascular smooth muscle precursor development by coordinating the ECM biophysical and biochemical properties.

7.
Exp Hematol ; 43(4): 300-8.e1, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25534203

RESUMEN

Two types of CCAAT-enhancer-binding protein α (C/EBPα) mutants are found in acute myeloid leukemia (AML) patients: N-terminal frame-shift mutants (C/EBPα-N(m)) generating p30 as a dominant form and C-terminal basic leucine zipper domain mutants (C/EBPα-C(m)). We have previously shown that C/EBPα-K304_R323dup belonging to C/EBPα-C(m), but not C/EBPα-T60fsX159 belonging to C/EBPα-N(m), alone induced AML in mouse bone marrow transplantation (BMT) models. Here we show that various C/EBPα-C(m) mutations have a similar, but not identical, potential in myeloid leukemogenesis. Notably, like C/EBPα-K304_R323dup, any type of C/EBPα-C(m) tested (C/EBPα-S299_K304dup, K313dup, or N321D) by itself induced AML, albeit with different latencies after BMT; C/EBPα-N321D induced AML with the shortest latency. By analyzing the gene expression profiles of C/EBPα-N321D- and mock-transduced c-kit(+)Sca-1(+)Lin(-) cells, we identified Csf1r as a gene downregulated by C/EBPα-N321D. In addition, leukemic cells expressing C/EBPα-C(m) exhibited low levels of colony stimulating factor 1 receptor in mice. On the other hand, transduction with C/EBPα-N(m) did not influence Csf1r expression in c-kit(+)Sca-1(+)Lin(-) cells, implying a unique role for C/EBPα-C(m) in downregulating Csf1r. Importantly, Csf1r overexpression collaborated with C/EBPα-N321D to induce fulminant AML with leukocytosis in mouse BMT models to a greater extent than did C/EBPα-N321D alone. Collectively, these results suggest that C/EBPα-C(m)-mediated downregulation of Csf1r has a negative, rather than a positive, impact on the progression of AML involving C/EBPα-C(m), which might possibly be accelerated by additional genetic and/or epigenetic alterations inducing Csf1r upregulation.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Regulación hacia Abajo/fisiología , Leucemia Mieloide Aguda/patología , Mutación , Receptor de Factor Estimulante de Colonias de Macrófagos/fisiología , Animales , Secuencia de Bases , Proteína alfa Potenciadora de Unión a CCAAT/genética , Cartilla de ADN , Progresión de la Enfermedad , Citometría de Flujo , Humanos , Leucemia Mieloide Aguda/fisiopatología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Proc Jpn Acad Ser B Phys Biol Sci ; 90(10): 389-404, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25504228

RESUMEN

Myeloid malignancies consist of acute myeloid leukemia (AML), myelodysplastic syndromes (MDS) and myeloproliferative neoplasm (MPN). The latter two diseases have preleukemic features and frequently evolve to AML. As with solid tumors, multiple mutations are required for leukemogenesis. A decade ago, these gene alterations were subdivided into two categories: class I mutations stimulating cell growth or inhibiting apoptosis; and class II mutations that hamper differentiation of hematopoietic cells. In mouse models, class I mutations such as the Bcr-Abl fusion kinase induce MPN by themselves and some class II mutations such as Runx1 mutations induce MDS. Combinations of class I and class II mutations induce AML in a variety of mouse models. Thus, it was postulated that hematopoietic cells whose differentiation is blocked by class II mutations would autonomously proliferate with class I mutations leading to the development of leukemia. Recent progress in high-speed sequencing has enabled efficient identification of novel mutations in a variety of molecules including epigenetic factors, splicing factors, signaling molecules and proteins in the cohesin complex; most of these are not categorized as either class I or class II mutations. The functional consequences of these mutations are now being extensively investigated. In this article, we will review the molecular basis of hematological malignancies, focusing on mouse models and the interfaces between these models and clinical findings, and revisit the classical class I/II hypothesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Epigénesis Genética , Neoplasias Hematológicas/genética , Leucemia Mieloide Aguda/genética , Mutación , Síndromes Mielodisplásicos/genética , Animales , Proliferación Celular/genética , Transformación Celular Neoplásica/metabolismo , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Neoplasias Hematológicas/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Ratones , Síndromes Mielodisplásicos/metabolismo
9.
Blood ; 123(25): 3932-42, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24825862

RESUMEN

High levels of HES1 expression are frequently found in BCR-ABL(+) chronic myelogenous leukemia in blast crisis (CML-BC). In mouse bone marrow transplantation (BMT) models, co-expression of BCR-ABL and Hes1 induces CML-BC-like disease; however, the underlying mechanism remained elusive. Here, based on gene expression analysis, we show that MMP-9 is upregulated by Hes1 in common myeloid progenitors (CMPs). Analysis of promoter activity demonstrated that Hes1 upregulated MMP-9 by activating NF-κB. Analysis of 20 samples from CML-BC patients showed that MMP-9 was highly expressed in three, with two exhibiting high levels of HES1 expression. Interestingly, MMP-9 deficiency impaired the cobblestone area-forming ability of CMPs expressing BCR-ABL and Hes1 that were in conjunction with a stromal cell layer. In addition, CMPs expressing BCR-ABL and Hes1 secreted MMP-9, promoting the release of soluble Kit-ligand (sKitL) from stromal cells, thereby enhancing proliferation of the leukemic cells. In accordance, mice transplanted with CMPs expressing BCR-ABL and Hes1 exhibited high levels of sKitL as well as MMP-9 in the serum. Importantly, MMP-9 deficiency impaired the development of CML-BC-like disease induced by BCR-ABL and Hes1 in mouse BMT models. The present results suggest that Hes1 promotes the development of CML-BC, partly through MMP-9 upregulation in leukemic cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Crisis Blástica/genética , Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Metaloproteinasa 9 de la Matriz/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Crisis Blástica/metabolismo , Trasplante de Médula Ósea/métodos , Movimiento Celular/genética , Proliferación Celular , Citometría de Flujo , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Estimación de Kaplan-Meier , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Genéticos , FN-kappa B/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor de Transcripción HES-1 , Regulación hacia Arriba
10.
Exp Hematol ; 42(5): 369-379.e3, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24486648

RESUMEN

We have previously shown that elevated expression of Hairy enhancer of split 1 (Hes1) contributes to blast crisis transition in Bcr-Abl-positive chronic myelogenous leukemia. Here we investigate whether Hes1 is involved in the development of other myeloid neoplasms. Notably, Hes1 expression was elevated in only a few cases of 65 samples with different types of myeloid neoplasms. Interestingly, elevated expression of Hes1 was found in two of five samples of Fip1-like1 platelet-derived growth factor receptor-α (FIP1L1-PDGFA)-positive myeloid neoplasms associated with eosinophilia. Whereas FIP1L1-PDGFRα alone induced acute T-cell leukemia or myeloproliferative neoplasms in mouse bone marrow transplantation models, mice transplanted with bone marrow cells expressing both Hes1 and FIP1L1-PDGFRα developed acute leukemia characterized by an expansion of myeloid blasts and leukemic cells without eosinophilic granules. FIP1L1-PDGFRα conferred cytokine-independent growth to Hes1-transduced common myeloid progenitors, interleukin-3-dependent cells. Imatinib inhibited the growth of common myeloid progenitors expressing Hes1 with FIP1L1-PDGFRα, but not with imatinib-resistant FIP1L1-PDGFRα mutants harboring T674I or D842V. In contrast, ponatinib efficiently eradicated leukemic cells expressing Hes1 and the imatinib-resistant FLP1L1-PDGFRΑ mutant in vitro and in vivo. Thus, we have established mouse models of FIP1L1-PDGFRA-positive leukemia in myeloid blast crisis, which will help elucidate the pathogenesis of the disease and develop a new treatment for it.


Asunto(s)
Antineoplásicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Crisis Blástica/metabolismo , Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Leucemia Mieloide Aguda/mortalidad , Neoplasias Experimentales/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Sustitución de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Benzamidas/farmacología , Crisis Blástica/genética , Crisis Blástica/patología , Femenino , Proteínas de Homeodominio/genética , Humanos , Mesilato de Imatinib , Interleucina-3/biosíntesis , Interleucina-3/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Mutación Missense , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas de Fusión Oncogénica/genética , Piperazinas/farmacología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Pirimidinas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Transcripción HES-1 , Factores de Escisión y Poliadenilación de ARNm/genética
11.
Int J Hematol ; 96(5): 638-48, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23097187

RESUMEN

Activating mutations of c-Kit are frequently found in acute myeloid leukemia (AML) patients harboring t(8;21) chromosomal translocation generating a fusion protein AML1-ETO. Here we show that an active mutant of c-Kit cooperates with AML1-ETO to induce AML in mouse bone marrow transplantation models. Leukemic cells expressing AML1-ETO with c-Kit(D814V) were serially transplantable. Transplantation experiments indicated that lineage(-)c-Kit(+)Sca-1(+) (KSL) leukemic cells, but not lineage(+) leukemic cells, were enriched for leukemia stem cells (LSCs). Comparison of gene expression profiles between KSL leukemic and normal cells delineated that CD200R1 was highly expressed in KSL leukemic cells as compared with KSL normal cells. Upregulation of CD200R1 was verified in lineage(-) leukemic cells, but not in lineage(+) leukemic cells. CD200R1 expression in the lineage(-) leukemic cells was not correlated with the frequency of LSCs, indicating that CD200R1 is not a useful marker for LSCs in these models. Interestingly, CD200R1 was upregulated in KSL cells transduced with AML1-ETO, but not with other leukemogenic mutants, including c-Kit(D814V), AML1(D171N), and AML1(S291fsX300). Consistently, upregulation of CD200R1 in lineage(-) leukemic cells was observed only in the BM of mice suffering from AML1-ETO-positive leukemia. In conclusion, AML1-ETO upregulated CD200R1 in lineage(-) cells, which was characteristic of AML1-ETO-positive leukemia in mice.


Asunto(s)
Antígenos de Superficie/biosíntesis , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Leucémica de la Expresión Génica , Leucemia/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Superficie Celular/biosíntesis , Factores de Transcripción/metabolismo , Regulación hacia Arriba , Sustitución de Aminoácidos , Animales , Antígenos de Superficie/genética , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Proteínas de Unión al ADN/genética , Leucemia/genética , Leucemia/patología , Ratones , Mutación Missense , Proteínas de Fusión Oncogénica/genética , Receptores de Orexina , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Ratas , Receptores de Superficie Celular/genética , Factores de Transcripción/genética
12.
Intern Med ; 48(2): 111-4, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19145056

RESUMEN

AIDS-related lymphoma (ARL) is a serious complication of HIV infection. We performed MEAM (MCNU + etoposide + cytarabine + L-PAM) regimen with autologous stem cell transplantation (ASCT) for three patients with refractory or relapsed ARL. All three patients had been treated with highly active anti-retroviral therapy (HAART) during the course of the treatment regimen and ASCT. The regimen was well tolerable, and no uncontrollable infection was noted. All patients are still alive and maintain complete remission at 24, 20 and 9 months after transplantation. ASCT using MEAM regimen as a conditioning regimen was feasible for our patients with refractory or relapsed ARL.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Linfoma Relacionado con SIDA/terapia , Acondicionamiento Pretrasplante/métodos , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Terapia Antirretroviral Altamente Activa , Citarabina/administración & dosificación , Etopósido/administración & dosificación , Femenino , Infecciones por VIH/tratamiento farmacológico , Humanos , Masculino , Melfalán/administración & dosificación , Compuestos de Nitrosourea/administración & dosificación , Recurrencia , Terapia Recuperativa , Acondicionamiento Pretrasplante/efectos adversos , Trasplante Autólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...