Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Journal of Antimicrobial Chemotherapy, Oxford University Press (OUP), Vol. 75, No. 5 ( 2020-05-01), p. 1324-1331
    Abstract: Safety data about rilpivirine use during pregnancy remain scarce, and rilpivirine plasma concentrations are reduced during second/third trimesters, with a potential risk of viral breakthroughs. Thus, French guidelines recommend switching to rilpivirine-free combinations (RFCs) during pregnancy. Objectives To describe the characteristics of women initiating pregnancy while on rilpivirine and to compare the outcomes for virologically suppressed subjects continuing rilpivirine until delivery versus switching to an RFC. Methods In the ANRS-EPF French Perinatal cohort, we included women on rilpivirine at conception in 2010–18. Pregnancy outcomes were compared between patients continuing versus interrupting rilpivirine. In women with documented viral suppression ( & lt;50 copies/mL) before 14 weeks of gestation (WG) while on rilpivirine, we compared the probability of viral rebound (≥50 copies/mL) during pregnancy between subjects continuing rilpivirine versus those switching to RFC. Results Among 247 women included, 88.7% had viral suppression at the beginning of pregnancy. Overall, 184 women (74.5%) switched to an RFC (mostly PI/ritonavir-based regimens) at a median gestational age of 8.0 WG. Plasma HIV-1 RNA nearest delivery was & lt;50 copies/mL in 95.6% of women. Among 69 women with documented viral suppression before 14 WG, the risk of viral rebound was higher when switching to RFCs than when continuing rilpivirine (20.0% versus 0.0%, P = 0.046). Delivery outcomes were similar between groups (overall birth defects, 3.8/100 live births; pregnancy losses, 2.0%; preterm deliveries, 10.6%). No HIV transmission occurred. Conclusions In virologically suppressed women initiating pregnancy, continuing rilpivirine was associated with better virological outcome than changing regimen. We did not observe a higher risk of adverse pregnancy outcomes.
    Type of Medium: Online Resource
    ISSN: 0305-7453 , 1460-2091
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2020
    detail.hit.zdb_id: 1467478-6
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3728-3728
    Abstract: Abstract 3728 The occurrence of JAK2-V617F mutation has been reported during the evolution of several patients with Ph1-positive CML. In all cases where a clonal analysis has been performed, the involvement of two different hematopoietic stem cells (HSC) has been demonstrated, with the presence of two different myeloproliferative disorders (MPDs). The occurrence of the V617F mutation in a leukemic stem cell (LSC) harboring the BCR-ABL rearrangement has not been reported so far. In a 63- year old patient with a diagnosis of high Sokal score Ph1+ CML, the initial therapy with Imatinib led to efficient cytoreduction and complete cytogenetic response but with persistent splenomegaly. Upon disease progression, he was treated successively with IFN-alpha, ARA-C, Dasatinib and Nilotinib with transient responses and persistent splenomegaly. The progressive increase of platelet counts led to the discovery of JAK2-V617F mutation, which appeared to be present from the diagnosis of CML with a progressive increase (40 % at diagnosis up to 70% at the time of the discovery of JAK2 mutation). To determine if BCR-ABL and JAK2 mutation were present in the same cells, a clonogenic assay was performed and single BFU-E's (n=11) were analyzed individually by q-RT-PCR. All 11 BFU-E expressed BCR-ABL at high levels with a mean BCR-ABL/ABL ratio of 157% (Range 91–207 %). The same individual BFU-E also revealed the presence of a heterozygous JAK2-V617F mutation. To generate a HSC model harboring simultaneously these two major molecular events, we have generated induced pluripotent stem cells (iPSC) from leukemic cells using Sendai-virus mediated Oct4, Sox2, Klf4 and c-Myc gene transfer. We have also generated iPSC from non-leukemic (BCR-ABL-negative, JAK2-V617F-negative) endothelial progenitor cells (EPC) obtained from peripheral blood. iPSC generated from both sources expressed pluripotency markers (Tra-1–60, SSEA-3, SSEA-4) and generated teratoma in NOD/SCID mice. In 24 individual leukemic iPSC clones which were amplified, Ph1-chromosome was found to be present in all mitoses and BCR-ABL was highly expressed (Mean ratio 140%). Each iPSC exhibited the same V617F mutation which co-existed in the primary leukemic BFU-E. None of the non-leukemic iPSC expressed BCR-ABL neither V617F. To determine the hematopoietic potential of both iPSC, we have generated day-16 embryoid bodies (EB) followed by induction of hematopoiesis. Day 16 EB's generated variable numbers of CD34+ cells (12–40 %, n=4) and clonogenic potential (25–340 CFC/105 D16-EB) with evidence of growth-factor-independent CFC's. Interestingly, IM and Dasatinib had partial inhibitory effects on CFC activity, but a combination of Pimozide (STAT5 inhibitor) and IM had a major synergistic effect. As neither normal iPSC nor human embryonic stem cells (hESC) are able to generate definitive, long-term hematopoiesis, we have tested the long-term HSC potential of these iPSC in long-term culture initiating cell (LTC-IC) assays as compared to human ESC line H1. In these conditions, leukemic iPSC generated hematopoietic cells with terminal granulocytic and erythroblastic differentiation and gave rise for the first time to an LTC-IC- derived progeny at 6 weeks (200 CFU-C/105 Day-16 EB) whereas in the same time H1-derived cells did not generate any significant LTC-IC potential (3 CFU-C/105 D-16 EB). Interestingly, non-leukemic iPSC generated also hematopoietic cells but exhibited a major genetic instability in culture with appearance after several passages, of a t (5;7) translocation and major deletions/duplications in CGH arrays. To determine a potential upstream molecular event which might be involved at the origin of the occurrence of BCR-ABL and JAK2 mutation in the same HSC, a whole genome sequencing strategy is underway. In conclusion, we report for the first time the occurrence of two major molecular events involved in CML and non-CML MPDs in the same LSC and demonstrate the utility of iPSC modeling to study the earliest HSC involved. These results suggest strongly that the co-existence of BCR-ABL and V617F enables the specification of iPSC towards hematopoieis which could lead to the identification of specific hematopoietic signaling pathways. Finally, clonal analysis of hematopoietic cells in CML patients with demonstration of Jak2V617 mutations is worthwhile to determine the prevalence of the simultaneous occurrence of these two molecular events in the same cell. Disclosures: Turhan: Novartis, Bristol Myers Squibb: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 514-514
    Abstract: Genetic instability is a hallmark of chronic myeloid leukemia (CML). Recently, several major abnormalities in DNA repair mechanisms have been identified in primitive CML cells that likely explain the additional mutations these cells develop leading to their selective growth under tyrosine kinase inhibitor (TKI) therapies. It seems likely that such mechanisms also underlie disease progression in CML. However, an understanding of the specific somatic mutations involved and investigations of their resulting effects on the biological behavior of primary sources of primitive chronic phase (CP) CML cells is extremely challenging. As an alternative approach, we have now explored the possibility of applying whole genome sequencing (WGS) to induced pluripotent stem cells (iPSCs) derived from primitive CML cells to determine if such iPSCs, genocopy the mutations present in the diagnostic sample from which they were generated and whether primitive hematopoietic cells derived from these iPSCs might be useful for future drug screening experiments. To this end, we chosen a CML patient whose CP clonogenic cells contained both the Ph1 chromosome and the JAK2 V617F mutation and whose disease progressed into an accelerated phase (AP) during TKI therapy. iPSC were generated from leukemic cells obtained at the time of AP using Oct4, Sox2, Klf4 and c-Myc gene transfer. The presence of both BCR-ABL and JAK2 V617F was confirmed in 24/24 iPSC colonies. A control iPSC line negative for both genes was similarly established from the patient’s CD34+CD31+ endothelial progenitors purified from peripheral blood. We then performed WGS on DNA prepared from the leukemic cells obtained at diagnosis of CP (CML 006), the AP cell-derived iPSCs (PB34), and the control non-leukemic iPSCs (PB13), using a HighSeq Illumina platform. WGS revealed 845,175 somatic SNVs and 68,817 somatic short Indels in the CP leukemic cells at diagnosis that were not present in the non-leukemic iPSCs (PB13). 49,225 of these SNVs and 11,665 of the short Indels were novel (absent in the dbSNP database), and 419 were found in the COSMIC database. We identified 274 novel SNVs (3 missense, 161 nonsense, 108 synonymous and 2 splice site mutations) and 46 short Indels (19 insertions and 27 deletions). Most of the novel coding SNVs and Indels were heterozygous and an estimation of the variant allele frequency indicated these were present in virtually all leukemic cells. In addition to the JAK2 V617F mutation that was present at diagnosis, we found a novel frame shift mutation in exon 12 of ASXL1 gene (p.S871YfsX5) leading to protein truncation, a genetic event that has also been associated with myeloproliferative neoplasms (MPNs) and AML. We also identified several novel SNVs predicted by SIFT, Provean and PolyPhen-2 algorithms to be deleterious for protein structure. These novel mutations were found in genes relevant for the pathophysiology of MPNs, including the catenin (CTNNA1 R204C, and AIDA K235T), RAS (RREB1 P789T), autophagy (ULK1 R553C) cellular antioxidant defense (GSR S293C), RNA nuclear transport (NUP160 start loss) pathways. Individual sequencing confirmed the presence of these mutations in PB34 and their absence in PB13 (non-leukemic iPSC). We next compared the sequence data from the AP leukemic cell-derived iPSCs (PB34) with the diagnostic data (CML006). This analysis showed only 799 additional somatic SNVs and 96 new short Indels compared with those already evident in the cells present at diagnosis. Only 4 (3 non synonymous and 1 synonymous) SNVs and no Indels were found in exons. These mutations could have appeared during the application of the reprogramming process to the AP leukemic cell-derived iPSCs; none was an obvious contributor to MPN pathophysiology. Finally, we showed that day16 embryoid bodies derived from the PB34 iPSCs contained expected numbers of CD34+ cells (18±11%, n=6) and BCR-ABL-expressing hematopoietic colony-forming cells (CFCs, 143±64 / 105 cells, n= 6). These CFCs showed a slight inhibitory response to imatinib (54±15% colonies obtained in 1 µM IM, n=4) whereas a combination of IM and Pimozide (a STAT5 phosphorylation inhibitor), reduced survival another ∼10-fold. In conclusion, we have provided proof-of-principle results illustrating the potential of iPSC technology in combination with WGS to dissect the clonal evolution of disease progression in CML and develop patient specific drug screens that could build on this data. Disclosures: Turhan: BMS, Novartis: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3726-3726
    Abstract: Abstract 3726 Recent data showed that BCR-ABL expressing leukemic stem cells (LSCs) persist in vivo in patients in complete molecular remission (CMR) according to European Leukemia Net criteria (Chomel et al, Blood 2011, Chu et al, Blood 2011). It has also been established that, in this category of patients (CMR 4.5– 5), Imatinib Mesylate (IM) discontinuation leads to relapse in the majority of cases (60%), especially during the first 6 months. Although very likely, a formal link has not been established between these two phenomena and, in particular, the prevalence of LSC persistence in a cohort of patients treated with TKI as a first line therapy has not been established. To determine the prevalence of LSC persistence in CMR induced by first line TKI therapies, we have evaluated by clonogenic and long-term culture initiating cell (LTC-IC) assays, the presence of BCR-ABL expressing LSCs in marrow samples of 26 patients in CMR for 〉 2 years. CMR was induced by IM (n=22), Dasatinib (n=3) or Nilotinib (n=1) as a first line treatment. CD34+ cells were isolated from bone marrow aspirates (2–4 ml) using immunomagnetic columns and a clonogenic assay was performed. At week+2, clonogenic progenitors were counted and 20 to 40 individual hematopoietic colonies were plucked from methylcellulose for RNA extraction. In some cases, pooled CFU-Cs were also analyzed (20 pools of 10 hematopoietic colonies). The same CD34+ marrow sample was used to start LTC-IC assays, which were performed in murine MS5 cell feeders. At week+6 the cultures were sacrificed and the number of LTC-IC-derived clonogenic cells was determined. Hematopoietic colonies were plucked as decribed above for CFU-Cs for RNA extraction and BCR-ABL expression was quantified by q-RT-PCR. For 4 patients, the yield of CD34+ cells was not sufficient for stem cell assays (3 pts treated with IM; 1 with Dasatinib). qRT-PCR analyses of CFU-Cs revealed the presence of BCR-ABL expressing cells in 3 patients out of 16 tested so far. In all three patients in CMR induced by IM, this involvement can be estimated to approximately 100–360 leukemic CFU-GM/ml of bone marrow. Concerning LTC-IC assays, 2 patients out of 6 tested so far had evidence of BCR-ABL expressing LTC-IC-derived progenitors, one of these patients having also involvement of the clonogenic compartment. In both patients, there was a significant involvement of LTC-IC-derived progeny estimated to 1000–4000 LTC-IC derived progenitor/ml of bone marrow. In the 4 patients in whom LTC-IC compartment was found to be negative for LSCs, CMR was induced by IM in 3 Pts (IM 400 mg n= 2, IM 800 mg for accelerated phase n=1) and by Dasatinib (100 mg) in 1 Pt. Interestingly, in all q-RT-PCR assays, the amount of BCR-ABL mRNA (evaluated by the BCR-ABL/ABL ratio) in individual CFU-Cs appeared to be low, confirming our previous results (Chomel et al Blood 2012) and suggesting that these cells are not addict to BCR-ABL-TK activity for their survival. In 12 patients, the evaluation of LTC-IC compartment is underway. Overall, These data reveal that in patients in CMR induced by IM as a first line therapy, our strategy could identify two types of stem cell response, with some patients presenting a complete eradication of the leukemic stem cell compartment (within the limits of the sensitivity of the test) and others showing high numbers of BCR-ABL expressing stem cells despite the continuous CMR status on TKI therapy. It remains to be determined if the involvement of LTC-IC-compartment (in 2/6 analyzed so far) by LSC expressing low levels of BCR-ABL can be overcome by long-term TKI therapy in some patients. In addition, it will be of interest to determine if a correlation exists between LSC-positive patients the potential occurence of a molecular relapse upon discontinuation of TKI therapy. From the mechanistic point of view, the persistence of residual marrow LSCs in some CML patients in CMR strongly suggests the presence of active mechanisms of either LSC retention and/or LSC quiescence-promoting interactions within in the hematopoietic niche. Finally, the comparison of the LSC persistence phenomenon in CMR induced by TKI2 versus IM as first line treatment will be of major interest to determine the LSC eradication potential of these drugs. Disclosures: Rea: Bristol Myers-Squibb, Novartis, and Teva: Honoraria. Turhan:Novartis, Bristol Myers Squibb: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: European Journal of Medical Genetics, Elsevier BV, Vol. 61, No. 6 ( 2018-06), p. 322-328
    Type of Medium: Online Resource
    ISSN: 1769-7212
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2018
    detail.hit.zdb_id: 2186601-6
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Endocrine Abstracts, Bioscientifica, ( 2022-05-07)
    Type of Medium: Online Resource
    ISSN: 1479-6848
    Language: Unknown
    Publisher: Bioscientifica
    Publication Date: 2022
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Endocrine Abstracts, Bioscientifica, ( 2023-05-02)
    Type of Medium: Online Resource
    ISSN: 1479-6848
    Language: Unknown
    Publisher: Bioscientifica
    Publication Date: 2023
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: The Lancet Diabetes & Endocrinology, Elsevier BV, Vol. 9, No. 12 ( 2021-12), p. 813-824
    Type of Medium: Online Resource
    ISSN: 2213-8587
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Journal of the Endocrine Society, The Endocrine Society, Vol. 6, No. Supplement_1 ( 2022-11-01), p. A81-A81
    Abstract: Primary bilateral macronodular adrenal hyperplasia (PBMAH) with glucose-dependent insulinotropic polypeptide (GIP)-dependent Cushing's syndrome is caused by ectopic expression of GIP receptor (GIPR) in the adrenal lesions. Such ectopic expression of GIPR was also reported in other endocrine neoplasm, notably in somatotroph pituitary adenomas from acromegalic patients with paradoxical increase of GH after oral glucose load, suggesting a common molecular pathogenesis. We aimed to identify the driver event responsible for GIP-dependent PBMAH with Cushing's syndrome and ectopic GIPR expression in somatotropinomas. Methods We conducted an international, multicenter, cohort study. We collected blood and adrenal samples from patients who had undergone unilateral or bilateral adrenalectomy for GIP-dependent PBMAH with Cushing's syndrome. Adrenal samples from patients with PBMAH and Cushing's syndrome without food-dependent cortisol production were used as controls. We further collected somatotropinoma specimens from acromegaly patients followed at two expert endocrine centers in France. Results 17 patients with familial or sporadic GIP-dependent PBMAH with Cushing's syndrome were studied. We identified germline heterozygous mutations in the lysine demethylase 1A (KDM1A) gene in all 17 patients. We further identified a recurrent deletion of the short arm of chromosome 1 harboring the KDM1A locus in the adrenal lesions of affected patients. None of the 25 patients in the control group had KDM1A germline or somatic alterations. Concomitant genetic inactivation of both KDM1A alleles resulted in loss of KDM1A expression in the adrenal lesions. RNA-sequencing revealed the global impact of KDM1A loss in adrenal tissue on gene transcription and identified differentially regulated genes including those encoding for GIPR and other G-Protein-Coupled Receptors that may be involved in adrenal tumorigenesis and regulation of steroidogenesis. In vitro pharmacologic inhibition, silencing and knock-out by CRISPR-Cas9 genome editing of KDM1A led to an increase in GIPR transcripts and protein in human adrenocortical H295R cells. Somatotropinoma samples from 78 patients with acromegaly were studied. 24% of these patients presented with a paradoxical rise of GH after oral glucose load and expressed ectopically GIP-receptor in their somatotropinoma. None of the somatotropinomas harbored KDM1A pathogenic variants, but those from patients with paradoxical GH response displayed a recurrent chromosome 1p loss. Discussion We identified germline inactivating KDM1A mutations and loss of heterozygosity as a genetic predisposition to GIP-dependent PBMAH with Cushing's syndrome following a tumor suppressor gene model of tumorigenesis. We currently perform genetic screening in first-degree relatives of patients with GIP-dependent PBMAH with Cushing's syndrome and clinical examination with biochemical testing in asymptomatic KDM1A variant carriers. We did not identify somatic KDM1A mutations in somatotropinomas expressing GIPR ectopically, however their recurrent 1p chromosome loss suggests that KDM1A haploinsufficiency may contribute to GIPR expression in those tumors. Presentation: Saturday, June 11, 2022 12:15 p.m. - 12:30 p.m.
    Type of Medium: Online Resource
    ISSN: 2472-1972
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2022
    detail.hit.zdb_id: 2881023-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Clinical Chemistry, Oxford University Press (OUP), Vol. 68, No. 2 ( 2022-02-01), p. 313-321
    Abstract: To date, the usage of Galaxy, an open-source bioinformatics platform, has been reported primarily in research. We report 5 years’ experience (2015 to 2020) with Galaxy in our hospital, as part of the “Assistance Publique–Hôpitaux de Paris” (AP-HP), to demonstrate its suitability for high-throughput sequencing (HTS) data analysis in a clinical laboratory setting. Methods Our Galaxy instance has been running since July 2015 and is used daily to study inherited diseases, cancer, and microbiology. For the molecular diagnosis of hereditary diseases, 6970 patients were analyzed with Galaxy (corresponding to a total of 7029 analyses). Results Using Galaxy, the time to process a batch of 23 samples—equivalent to a targeted DNA sequencing MiSeq run—from raw data to an annotated variant call file was generally less than 2 h for panels between 1 and 500 kb. Over 5 years, we only restarted the server twice for hardware maintenance and did not experience any significant troubles, demonstrating the robustness of our Galaxy installation in conjunction with HTCondor as a job scheduler and a PostgreSQL database. The quality of our targeted exome sequencing method was externally evaluated annually by the European Molecular Genetics Quality Network (EMQN). Sensitivity was mean (SD)% 99 (2)% for single nucleotide variants and 93 (9)% for small insertion-deletions. Conclusion Our experience with Galaxy demonstrates it to be a suitable platform for HTS data analysis with vast potential to benefit patient care in a clinical laboratory setting.
    Type of Medium: Online Resource
    ISSN: 0009-9147 , 1530-8561
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2022
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages