16 research outputs found

    Detection of leukemic stem cell (CD26+) in patients with chronic myeloid leukemia with different molecular response

    Get PDF
    La leucemia mieloide crónica (LMC) se caracteriza por la t(9;22)(q34;q11), generando el gen de fusión BCR-ABL1 que codifica la oncoproteina P210 con actividad constitutiva de tirosina kinasa. Los pa-cientes que presentan una profunda y sostenida res-puesta molecular a los inhibidores de tirosina kinasa (ITK) pueden interrumpir el tratamiento. Sin em-bargo, aproximadamente el 50% de los casos presen-tan recurrencia molecular, probablemente debido a la persistencia de la stem cell leucémica (SCL) quies-cente (no replicativa, transcripcionalmente silente). Recientes publicaciones han demostrado que la ex-presión de la enzima dipeptidil peptidasa IV (CD26) está restringida a la fracción CD45+/CD34+/CD38- de la SCL en LMC y no se ha detectado en otras SCL mieloides/linfoides ni en medula ósea normal. Por esta razón CD26 es considerado un nuevo y especí-fico bio-marcador de LMC.El objetivo del trabajo fue detectar las SCL CD26+ en pacientes con LMC con diferente respuesta molecular (RM) y determinar si estas células persisten aun en casos con respuesta molecular profunda (RMP).Se analizaron 193 muestras de pacientes con LMC (107 sexo masculino y 86 femenino) para evaluar la SCL mediante citometría de flujo usando el panel de anticuerpos monoclonales: CD45, CD34, CD38, CD26, CD117, CD123, CD3 y HLA-DR. En para-lelo se realizó el estudio de la respuesta molecular mediante qRT-PCR BCR-ABL1 (Método Taqman). Ambos estudios se realizaron en simultáneo en la misma muestra, durante el seguimiento en diferen-tes momentos bajo tratamiento con ITK (imatinib, nilotinib o dasatinib). Los pacientes con una reducción de BCR-ABL1 ≥ a 3 log tenían una significativa menor proporción de casos con SCL CD26+ comparado con aquellos que tenían <3 log de reducción de los transcriptos (p<0.0003, OR: 3.4, 95% CI: 1,7 - 6,8). Consideran-do los 76 casos con RMP (33 RM4.0; 38 RM4.5 y 5 RM5.0), solamente 12/76 (16%) mostraron per-sistencia de la SCL CD26+. La presencia de la SCL CD26+ se redujo acorde aumenta la profundidad de la RM: 21%, 13% y 0% en RM4.0, RM4.5 y RM5.0 respectivamente. Nuestros resultados muestran que los pacientes con buena RM (≥3log), se asociaron con baja propor-ción de casos con SCL CD26+. Cuando la detección de SCL se evaluó exclusivamente en los casos con RMP, se observó que el decrecimiento de la SCL se asoció a mayor profundidad de la RM. La stem cell leucémica es altamente quiescente por lo cual podría estar presente aun en casos con respuesta molecular indetectable. En nuestro estudio la persistencia de SCL fue del 16% en casos con respuesta molecular profunda, indicando que la SCL persiste a pesar de la RM alcanzada. Este nuevo abordaje investigando la SCL podría ser útil en el seguimiento a largo plazo y de gran importancia en la evaluación de la recu-rrencia molecular en los casos incluidos en protoco-los de discontinuación.Chronic Myeloid Leukemia (CML) is characterized by the reciprocal translocation t(9;22)(q34;q21) resulting in the BCR-ABL1 fusion gene encoding the P210 oncoprotein with a constitutive tyrosine kinase (TK) activity. It is known that patients with at least two years in deep and sustained molecular response could stop TK inhibitor (TKI) treatment. However, half of them show molecular recurrence, probably due to the persistence of transcriptionally quiescent leukemic stem cells (LSC). Recent studies show that the expression of the enzyme dipeptidylpeptidase IV (CD26) is mainly restricted to the CD45+/CD34+/ CD38- fraction in CML LSC, and it is not found in other myeloid/lymphoid LSC or in normal bone marrow. For this reason, CD26 is considered a novel specific biomarker in CML. The aim of this study was to detect the CD26+ LSC in CML patients with different molecular responses (MR) and to assess if these cells remain even in deep molecular response (DMR). We have evaluated 193 CML patients (107 males and 86 females) for detection of LSC by flow cytometry using the panel: CD45, CD34, CD38, CD26, CD117, CD123, CD3 and HLA-DR and the BCR-ABL1 quantification by qRT-PCR (Taqman method). Both studies were carried out simultaneously on the same sample, during the follow up at different time points under TKI treatment (Imatinib, Nilotinib, Dasatinib). Patients with ≥ 3 BCR-ABL1 log reduction had a significantly lower percentage of cases with CD26+ LSC compared with those who had < 3 log reduction (p<0.0003, OR: 3.4, 95% CI: 1,7 - 6,8). Out of the 76 patients with DMR (33 in MR4.0, 38 in MR4.5 and 5 in MR5.0) only 12/76 (16%) showed persistence of CD26+ LSC. Furthermore, the presence of CD26+ LSC decreased accordingly to the achieved DMR: 21%, 13% and 0% in MR4.0, MR4.5 and MR5.0 respectively, without significant differences. Our results show that patients with good MR (≥3log) were significantly associated with a lower proportion of cases with LSC presence. When the LSC analysis was performed exclusively in cases with DMR, we observed that the decrease of LSC accompanied the deepness of the molecular response. Since the LSC is highly quiescent, it could be present even in cases with undetectable MR. In our study persistence of LSC in cases with DMR was 16%, indicating that these cells remain despite the MR achieved. This new approach to the study of the LSC could be useful in long-term follow-up and of great importance in the evaluation of molecular recurrence in cases included in discontinuation protocols.Fil: Bengio, R. M.. Academia Nacional de Medicina de Buenos Aires. Instituto de Investigaciones Hematológicas "Mariano R. Castex"; ArgentinaFil: Peña, M.. Academia Nacional de Medicina de Buenos Aires. Instituto de Investigaciones Hematológicas "Mariano R. Castex"; ArgentinaFil: Palacios, F.. Academia Nacional de Medicina de Buenos Aires. Instituto de Investigaciones Hematológicas "Mariano R. Castex"; ArgentinaFil: Moiraghi, B.. Gobierno de la Ciudad de Buenos Aires. Hospital General de Agudos "Ramos Mejía"; ArgentinaFil: Negri Aranguren, P.. Instituto Privado de Hematologia y Hemoterapia; ArgentinaFil: Enrico, A.. Hospital Italiano de La Plata; ArgentinaFil: Mariano, R.. Provincia de Entre Rios. Hospital San Martin; ArgentinaFil: Toloza, Maria Jazmin Ayelen. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Medicina Experimental. Academia Nacional de Medicina de Buenos Aires. Instituto de Medicina Experimental; ArgentinaFil: Larripa, Irene Beatriz. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Medicina Experimental. Academia Nacional de Medicina de Buenos Aires. Instituto de Medicina Experimental; Argentin

    Eighteen Years of Molecular Genotyping the Hemophilia Inversion Hotspot: From Southern Blot to Inverse Shifting-PCR

    Get PDF
    The factor VIII gene (F8) intron 22 inversion (Inv22) is a paradigmatic duplicon-mediated rearrangement, found in about one half of patients with severe hemophilia A worldwide. The identification of this prevalent cause of hemophilia was delayed for nine years after the F8 characterization in 1984. The aim of this review is to present the wide diversity of practical approaches that have been developed for genotyping the Inv22 (and related int22h rearrangements) since discovery in 1993. The sequence— Southern blot, long distance-PCR and inverse shifting-PCR—for Inv22 genotyping is an interesting example of scientific ingenuity and evolution in order to resolve challenging molecular diagnostic problems

    Topoisomerase II-Mediated DNA Damage Is Differently Repaired during the Cell Cycle by Non-Homologous End Joining and Homologous Recombination

    Get PDF
    Topoisomerase II (Top2) is a nuclear enzyme involved in several metabolic processes of DNA. Chemotherapy agents that poison Top2 are known to induce persistent protein-mediated DNA double strand breaks (DSB). In this report, by using knock down experiments, we demonstrated that Top2α was largely responsible for the induction of γH2AX and cytotoxicity by the Top2 poisons idarubicin and etoposide in normal human cells. As DSB resulting from Top2 poisons-mediated damage may be repaired by non-homologous end joining (NHEJ) or homologous recombination (HR), we aimed to analyze both DNA repair pathways. We found that DNA-PKcs was rapidly activated in human cells, as evidenced by autophosphorylation at serine 2056, following Top2-mediated DNA damage. The chemical inhibition of DNA-PKcs by wortmannin and vanillin resulted in an increased accumulation of DNA DSB, as evaluated by the comet assay. This was supported by a hypersensitive phenotype to Top2 poisons of Ku80- and DNA-PKcs- defective Chinese hamster cell lines. We also showed that Rad51 protein levels, Rad51 foci formation and sister chromatid exchanges were increased in human cells following Top2-mediated DNA damage. In support, BRCA2- and Rad51C- defective Chinese hamster cells displayed hypersensitivity to Top2 poisons. The analysis by immunofluorescence of the DNA DSB repair response in synchronized human cell cultures revealed activation of DNA-PKcs throughout the cell cycle and Rad51 foci formation in S and late S/G2 cells. Additionally, we found an increase of DNA-PKcs-mediated residual repair events, but not Rad51 residual foci, into micronucleated and apoptotic cells. Therefore, we conclude that in human cells both NHEJ and HR are required, with cell cycle stage specificity, for the repair of Top2-mediated reversible DNA damage. Moreover, NHEJ-mediated residual repair events are more frequently associated to irreversibly damaged cells

    High cell-free DNA is associated with disease progression, inflammasome activation and elevated levels of inflammasome-related cytokine IL-18 in patients with myelofibrosis

    Get PDF
    Myelofibrosis (MF) is a clonal hematopoietic stem cell disorder classified among chronic myeloproliferative neoplasms, characterized by exacerbated myeloid and megakaryocytic proliferation and bone marrow fibrosis. It is induced by driver (JAK2/CALR/MPL) and high molecular risk mutations coupled to a sustained inflammatory state that contributes to disease pathogenesis. Patient outcome is determined by stratification into risk groups and refinement of current prognostic systems may help individualize treatment decisions. Circulating cell-free (cf)DNA comprises short fragments of double-stranded DNA, which promotes inflammation by stimulating several pathways, including inflammasome activation, which is responsible for IL-1β and IL-18 maturation and release. In this work, we assessed the contribution of cfDNA as a marker of disease progression and mediator of inflammation in MF. cfDNA was increased in MF patients and higher levels were associated with adverse clinical outcome, a high-risk molecular profile, advanced disease stages and inferior overall survival, indicating its potential value as a prognostic marker. Cell-free DNA levels correlated with tumor burden parameters and markers of systemic inflammation. To mimic the effects of cfDNA, monocytes were stimulated with poly(dA:dT), a synthetic double-stranded DNA. Following stimulation, patient monocytes released higher amounts of inflammasome-processed cytokine, IL-18 to the culture supernatant, reflecting enhanced inflammasome function. Despite overexpression of cytosolic DNA inflammasome sensor AIM2, IL-18 release from MF monocytes was shown to rely mainly on the NLRP3 inflammasome, as it was prevented by NLRP3-specific inhibitor MCC950. Circulating IL-18 levels were increased in MF plasma, reflecting in vivo inflammasome activation, and highlighting the previously unrecognized involvement of this cytokine in MF cytokine network. Monocyte counts were higher in patients and showed a trend towards correlation with IL-18 levels, suggesting monocytes represent a source of circulating IL-18. The close correlation shown between IL-18 and cfDNA levels, together with the finding of enhanced DNA-triggered IL-18 release from monocytes, suggest that cfDNA promotes inflammation, at least in part, through inflammasome activation. This work highlights cfDNA, the inflammasome and IL-18 as additional players in the complex inflammatory circuit that fosters MF progression, potentially providing new therapeutic targets

    Clinical activity of ponatinib in one patient with chronic myeloid leukemia in chronic phase with e19a2 transcript and T315I mutation

    Get PDF
    Background: Chronic myeloid leukemia (CML) is a hematological disorder that in rare cases, mainly in CML neutrophilic, presents the e19a2 rearrangement. The encoded product is a 230-KDa protein. Despite the remarkable responses to treatment of most patients, a small but significant fraction of them develop clinical resistance to the tyrosine kinase inhibitors (TKIs). The most common mechanism of resistance is point mutations in the ABL1 kinase domain. The recently approved third-generation TKI ponatinib demonstrated remarkable activity in patients with multi-TKI-resistant disease. Particularly impressive was its efficacy in patients with T315I mutation that is resistant to all other TKIs. Methods: Qualitative PCR was carried out by multiplex approach. Relative transcripts quantification was performed by one-step realtime PCR, with a specific Taqman probe and primers for the e19a2 rearrangement. We carried out a mutational screening by high-resolution melting, and the mutation was identified by Sanger method. The mutation burden was quantified by quantitative PCR using allele-specific primers. Results: In a patient with CML, we identified a PCR product corresponding to e19a2 rearrangement harboring T315I mutation. At the time of mutational analysis, during dasatinib treatment, the T315I clone was 100% and the quantification of BCR-ABL1 was 18%. After ponatinib therapy, the T315I mutation burden decreased down to undetectable levels and the BCR-ABL1 transcripts showed a very low value (0.011%). Conclusions: Here, we report the hematological, cytogenetic, and molecular response of a patient with refractory CML in chronic phase with e19a2 transcripts, carrying T315I mutation that was successfully treated with ponatinib.Fil: Ferri, Cristian Alberto. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Medicina Experimental. Academia Nacional de Medicina de Buenos Aires. Instituto de Medicina Experimental; ArgentinaFil: Bianchini, Michele. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Medicina Experimental. Academia Nacional de Medicina de Buenos Aires. Instituto de Medicina Experimental; ArgentinaFil: Bengió, Raquel M.. Academia Nacional de Medicina de Buenos Aires. Instituto de Investigaciones Hematológicas "Mariano R. Castex"; ArgentinaFil: Moiraghi, Elena B.. Gobierno de la Ciudad de Buenos Aires. Hospital General de Agudos "Ramos Mejía"; ArgentinaFil: Gonzalez, Mariana Selena. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Medicina Experimental. Academia Nacional de Medicina de Buenos Aires. Instituto de Medicina Experimental; ArgentinaFil: Noriega, Maria Fernanda. Academia Nacional de Medicina de Buenos Aires. Instituto de Investigaciones Hematológicas "Mariano R. Castex"; ArgentinaFil: Larripa, Irene Beatriz. Academia Nacional de Medicina de Buenos Aires. Instituto de Investigaciones Hematológicas "Mariano R. Castex"; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Medicina Experimental. Academia Nacional de Medicina de Buenos Aires. Instituto de Medicina Experimental; Argentin
    corecore