- Medical Biology - Research Publications
Medical Biology - Research Publications
Permanent URI for this collection
Search Results
Now showing
1 - 10 of 587
-
ItemCITED2 coordinates key hematopoietic regulatory pathways to maintain the HSC pool in both steady-state hematopoiesis and transplantation.Lawson, H ; van de Lagemaat, LN ; Barile, M ; Tavosanis, A ; Durko, J ; Villacreces, A ; Bellani, A ; Mapperley, C ; Georges, E ; Martins-Costa, C ; Sepulveda, C ; Allen, L ; Campos, J ; Campbell, KJ ; O'Carroll, D ; Göttgens, B ; Cory, S ; Rodrigues, NP ; Guitart, AV ; Kranc, KR (Elsevier BV, 2021-11-09)Hematopoietic stem cells (HSCs) reside at the apex of the hematopoietic differentiation hierarchy and sustain multilineage hematopoiesis. Here, we show that the transcriptional regulator CITED2 is essential for life-long HSC maintenance. While hematopoietic-specific Cited2 deletion has a minor impact on steady-state hematopoiesis, Cited2-deficient HSCs are severely depleted in young mice and fail to expand upon aging. Moreover, although they home normally to the bone marrow, they fail to reconstitute hematopoiesis upon transplantation. Mechanistically, CITED2 is required for expression of key HSC regulators, including GATA2, MCL-1, and PTEN. Hematopoietic-specific expression of anti-apoptotic MCL-1 partially rescues the Cited2-deficient HSC pool and restores their reconstitution potential. To interrogate the Cited2→Pten pathway in HSCs, we generated Cited2;Pten compound heterozygous mice, which had a decreased number of HSCs that failed to reconstitute the HSC compartment. In addition, CITED2 represses multiple pathways whose elevated activity causes HSC exhaustion. Thus, CITED2 promotes pathways necessary for HSC maintenance and suppresses those detrimental to HSC integrity.
-
ItemCutting edge approaches to detecting brain mosaicism associated with common focal epilepsies: implications for diagnosis and potential therapiesYe, Z ; Bennett, MF ; Bahlo, M ; Scheffer, IE ; Berkovic, SF ; Perucca, P ; Hildebrand, MS (TAYLOR & FRANCIS LTD, 2021-09-24)INTRODUCTION: Mosaic variants arising in brain tissue are increasingly being recognized as a hidden cause of focal epilepsy. This knowledge gain has been driven by new, highly sensitive genetic technologies and genome-wide analysis of brain tissue from surgical resection or autopsy in a small proportion of patients with focal epilepsy. Recently reported novel strategies to detect mosaic variants limited to brain have exploited trace brain DNA obtained from cerebrospinal fluid liquid biopsies or stereo-electroencephalography electrodes. AREAS COVERED: The authors review the data on these innovative approaches published in PubMed before 12 June 2021, discuss the challenges associated with their application, and describe how they are likely to improve detection of mosaic variants to provide new molecular diagnoses and therapeutic targets for focal epilepsy, with potential utility in other nonmalignant neurological disorders. EXPERT OPINION: These cutting-edge approaches may reveal the hidden genetic etiology of focal epilepsies and provide guidance for precision medicine.
-
ItemGerms and germlines: how "public" B-cell clones evolve in the gutJames, KR ; King, HW (WILEY, 2020-05-16)Chen et al. describe how B-cell clones observed in the gut of many different individuals (recurrent or "public" clonotypes) are shaped by the combined influences of common microbial antigens and underlying genomic recombination biases.
-
ItemBhlhe40: Gatekeeper of the GCNutt, SL ; Tellier, J (ROCKEFELLER UNIV PRESS, 2021-12-23)The generation of high-affinity antibodies in the germinal center (GC) requires interplay between GC B cells and T follicular helper cells. Rauschmeier et al. (2021. J. Exp. Med.https://doi.org/10.1084/jem.20211406) report that Bhlhe40 restrains GC output through distinct regulatory roles in both arms of the response.
-
ItemEfficient detection and classification of epigenomic changes under multiple conditionsBaldoni, PL ; Rashid, NU ; Ibrahim, JG (WILEY, 2021-05-03)Epigenomics, the study of the human genome and its interactions with proteins and other cellular elements, has become of significant interest in recent years. Such interactions have been shown to regulate essential cellular functions and are associated with multiple complex diseases. Therefore, understanding how these interactions may change across conditions is central in biomedical research. Chromatin immunoprecipitation followed by massively parallel sequencing (ChIP-seq) is one of several techniques to detect local changes in epigenomic activity (peaks). However, existing methods for differential peak calling are not optimized for the diversity in ChIP-seq signal profiles, are limited to the analysis of two conditions, or cannot classify specific patterns of differential change when multiple patterns exist. To address these limitations, we present a flexible and efficient method for the detection of differential epigenomic activity across multiple conditions. We utilize data from the ENCODE Consortium and show that the presented method, epigraHMM, exhibits superior performance to current tools and it is among the fastest algorithms available, while allowing the classification of combinatorial patterns of differential epigenomic activity and the characterization of chromatin regulatory states.
-
ItemOutcomes for Australian children with relapsed/refractory acute lymphoblastic leukaemia treated with blinatumomab.Sutton, R ; Pozza, LD ; Khaw, SL ; Fraser, C ; Revesz, T ; Chamberlain, J ; Mitchell, R ; Trahair, TN ; Bateman, CM ; Venn, NC ; Law, T ; Ong, E ; Heatley, SL ; McClure, BJ ; Meyer, C ; Marschalek, R ; Henderson, MJ ; Cross, S ; White, DL ; Kotecha, RS (Wiley, 2021-05)We report on the Australian experience of blinatumomab for treatment of 24 children with relapsed/refractory precursor B-cell acute lymphoblastic leukaemia (B-ALL) and high-risk genetics, resulting in a minimal residual disease (MRD) response rate of 58%, 2-year progression-free survival (PFS) of 39% and 2-year overall survival of 63%. In total, 83% (n = 20/24) proceeded to haematopoietic stem cell transplant, directly after blinatumomab (n = 12) or following additional salvage therapy (n = 8). Four patients successfully received CD19-directed chimeric antigen receptor T-cell therapy despite prior blinatumomab exposure. Inferior 2-year PFS was associated with MRD positivity (20%, n = 15) and in KMT2A-rearranged infants (15%, n = 9). Our findings highlight that not all children with relapsed/refractory B-ALL respond to blinatumomab and factors such as blast genotype may affect prognosis.
-
ItemNo Preview AvailableBlood and immune development in human fetal bone marrow and Down syndromeJardine, L ; Webb, S ; Goh, I ; Londono, MQ ; Reynolds, G ; Mather, M ; Olabi, B ; Stephenson, E ; Botting, RA ; Horsfall, D ; Engelbert, J ; Maunder, D ; Mende, N ; Murnane, C ; Dann, E ; McGrath, J ; King, H ; Kucinski, I ; Queen, R ; Carey, CD ; Shrubsole, C ; Poyner, E ; Acres, M ; Jones, C ; Ness, T ; Coulthard, R ; Elliott, N ; O'Byrne, S ; Haltalli, MLR ; Lawrence, JE ; Lisgo, S ; Balogh, P ; Meyer, KB ; Prigmore, E ; Ambridge, K ; Jain, MS ; Efremova, M ; Pickard, K ; Creasey, T ; Bacardit, J ; Henderson, D ; Coxhead, J ; Filby, A ; Hussain, R ; Dixon, D ; McDonald, D ; Popescu, D-M ; Kowalczyk, MS ; Li, B ; Ashenberg, O ; Tabaka, M ; Dionne, D ; Tickle, TL ; Slyper, M ; Rozenblatt-Rosen, O ; Regev, A ; Behjati, S ; Laurenti, E ; Wilson, NK ; Roy, A ; Goettgens, B ; Roberts, I ; Teichmann, SA ; Haniffa, M (NATURE PORTFOLIO, 2021-09-29)Haematopoiesis in the bone marrow (BM) maintains blood and immune cell production throughout postnatal life. Haematopoiesis first emerges in human BM at 11-12 weeks after conception1,2, yet almost nothing is known about how fetal BM (FBM) evolves to meet the highly specialized needs of the fetus and newborn. Here we detail the development of FBM, including stroma, using multi-omic assessment of mRNA and multiplexed protein epitope expression. We find that the full blood and immune cell repertoire is established in FBM in a short time window of 6-7 weeks early in the second trimester. FBM promotes rapid and extensive diversification of myeloid cells, with granulocytes, eosinophils and dendritic cell subsets emerging for the first time. The substantial expansion of B lymphocytes in FBM contrasts with fetal liver at the same gestational age. Haematopoietic progenitors from fetal liver, FBM and cord blood exhibit transcriptional and functional differences that contribute to tissue-specific identity and cellular diversification. Endothelial cell types form distinct vascular structures that we show are regionally compartmentalized within FBM. Finally, we reveal selective disruption of B lymphocyte, erythroid and myeloid development owing to a cell-intrinsic differentiation bias as well as extrinsic regulation through an altered microenvironment in Down syndrome (trisomy 21).
-
ItemNo Preview AvailableEnasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia (AG221-AML-005): a single-arm, phase 1b and randomised, phase 2 trialDiNardo, CD ; Schuh, AC ; Stein, EM ; Montesinos, P ; Wei, AH ; de Botton, S ; Zeidan, AM ; Fathi, AT ; Kantarjian, HM ; Bennett, JM ; Frattini, MG ; Martin-Regueira, P ; Lersch, F ; Gong, J ; Hasan, M ; Vyas, P ; Doehner, H (ELSEVIER SCIENCE INC, 2021-11-01)BACKGROUND: Enasidenib is an oral inhibitor of mutant isocitrate dehydrogenase-2 (IDH2) proteins. We evaluated the safety and activity of enasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia ineligible for intensive chemotherapy. METHODS: This open-label, phase 1b/2 trial was done at 43 clinical sites in 12 countries (the USA, Germany, Canada, the UK, France, Spain, Australia, Italy, the Netherlands, Portugal, Switzerland, and South Korea). Eligible patients were aged 18 years or older and had newly diagnosed, mutant-IDH2 acute myeloid leukaemia, and an Eastern Cooperative Oncology Group performance status of 0-2. In the phase 1b dose-finding portion, patients received oral enasidenib 100 mg/day or 200 mg/day in continuous 28-day cycles, plus subcutaneous azacitidine 75 mg/m2 per day for 7 days of each cycle. In phase 2, patients were randomly assigned (2:1) via an interactive web response system to enasidenib plus azacitidine or azacitidine-only, stratified by acute myeloid leukaemia subtype (de novo or secondary). The primary endpoint in the phase 2 portion was the overall response rate in the intention-to-treat population at a prespecified interim analysis (Aug 20, 2019) when all patients had at least 1 year of follow-up. Safety was assessed in all patients who received at least one dose of study drug. The trial is registered with ClinicalTrials.gov, NCT02677922, and is ongoing. FINDINGS: Between June 3, 2016, and Aug 2, 2018, 322 patients were screened and 107 patients with mutant-IDH2 acute myeloid leukaemia were enrolled. At data cutoff for the interim analysis, 24 patients (including two from the phase 1 portion) were still receiving their assigned treatment. Six patients were enrolled in the phase 1b dose-finding portion of the trial and received enasidenib 100 mg (n=3) or 200 mg (n=3) in combination with azacitidine. No dose-limiting toxicities occurred and the enasidenib 100 mg dose was selected for phase 2. In phase 2, 101 patients were randomly assigned to enasidenib plus azacitidine (n=68) or azacitidine only (n=33). Median age was 75 years (IQR 71-78). 50 (74%; 95% CI 61-84) patients in the enasidenib plus azacitidine combination group and 12 (36%; 20-55) patients in the azacitidine monotherapy group achieved an overall response (odds ratio 4·9 [95% CI 2·0-11·9]; p=0·0003). Common treatment-related grade 3 or 4 adverse events with enasidenib plus azacitidine were thrombocytopenia (25 [37%] of 68 vs six [19%] of 32 in the azacitidine-only group), neutropenia (25 [37%] vs eight [25%]), anaemia (13 [19%] vs seven [22%]), and febrile neutropenia (11 [16%] vs five [16%]). Serious treatment-related adverse events were reported in 29 (43%) patients in the combination group and 14 (44%) patients in the azacitidine-only group; serious treatment-related adverse events occurring in more than 5% of patients in either group were febrile neutropenia (nine [13%] in the combination group vs five [16%] in the azacitidine-only group), differentiation syndrome (seven [10%] vs none), and pneumonia (three [4%] vs two [6%]). No treatment-related deaths were reported. INTERPRETATION: Combination enasidenib plus azacitidine was well tolerated and significantly improved overall response rates compared with azacitidine monotherapy, suggesting that this regimen can improve outcomes for patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia. FUNDING: Bristol Myers Squibb.
-
ItemNo Preview AvailableMidostaurin reduces relapse in FLT3-mutant acute myeloid leukemia: the Alliance CALGB 10603/RATIFY trialLarson, RA ; Mandrekar, SJ ; Huebner, LJ ; Sanford, BL ; Laumann, K ; Geyer, S ; Bloomfield, CD ; Thiede, C ; Prior, TW ; Dohner, K ; Marcucci, G ; Voso, MT ; Klisovic, RB ; Galinsky, I ; Wei, AH ; Sierra, J ; Sanz, MA ; Brandwein, JM ; de Witte, T ; Niederwieser, D ; Appelbaum, FR ; Medeiros, BC ; Tallman, MS ; Krauter, J ; Schlenk, RF ; Ganser, A ; Serve, H ; Ehninger, G ; Amadori, S ; Gathmann, I ; Dohner, H ; Stone, RM (SPRINGERNATURE, 2021-03-02)The prospective randomized, placebo-controlled CALGB 10603/RATIFY trial (Alliance) demonstrated a statistically significant overall survival benefit from the addition of midostaurin to standard frontline chemotherapy in a genotypically-defined subgroup of 717 patients with FLT3-mutant acute myeloid leukemia (AML). The risk of death was reduced by 22% on the midostaurin-containing arm. In this post hoc analysis, we analyzed the cumulative incidence of relapse (CIR) on this study and also evaluated the impact of 12 4-week cycles of maintenance therapy. CIR analyses treated relapses and AML deaths as events, deaths from other causes as competing risks, and survivors in remission were censored. CIR was improved on the midostaurin arm (HR = 0.71 (95% CI, 0.54-0.93); p = 0.01), both overall and within European LeukemiaNet 2017 risk classification subsets when post-transplant events were considered in the analysis as events. However, when transplantation was considered as a competing risk, there was overall no significant difference between the risks of relapse on the two randomized arms. Patients still in remission after consolidation with high-dose cytarabine entered the maintenance phase, continuing with either midostaurin or placebo. Analyses were inconclusive in quantifying the impact of the maintenance phase on the overall outcome. In summary, midostaurin reduces the CIR.
-
ItemNo Preview AvailableOral azacitidine preserves favorable level of fatigue and health-related quality of life for patients with acute myeloid leukemia in remission: results from the phase III, placebo-controlled QUAZAR AML-001 trialRoboz, GJ ; Doehner, H ; Pocock, C ; Dombret, H ; Ravandi, F ; Jang, JH ; Selleslag, D ; Mayer, JR ; Martens, UM ; Liesveld, J ; Bernal, T ; Wang, MC ; Yu, P ; Shi, L ; Guo, S ; La Torre, I ; Skikne, B ; Dong, Q ; Braverman, J ; Nehme, SA ; Beach, CL ; Wei, AH (FERRATA STORTI FOUNDATION, 2021-12-01)Not available.