Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: PLOS Computational Biology, Public Library of Science (PLoS), Vol. 11, No. 7 ( 2015-7-9), p. e1004274-
    Type of Medium: Online Resource
    ISSN: 1553-7358
    Language: English
    Publisher: Public Library of Science (PLoS)
    Publication Date: 2015
    detail.hit.zdb_id: 2193340-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 22_Supplement_2 ( 2015-11-15), p. PR03-PR03
    Abstract: Acute myeloid leukemia is heterogeneous with respect to clinical outcome and molecular pathogenesis. Approximately 20% of AML cases are refractory to induction chemotherapy, and about 50% of patients ultimately relapse within a time interval that ranges from months to years. At the molecular level, diverse chromosomal abnormalities and genetic mutations have been observed across patients1. Although several clinical factors (age, white blood cell count), cytogenetic aberrations (t[15;17] translocation, loss of chromosome 5) 2-4, and genetic mutations (DNMT3A, FLT3) have been associated with differences in survival 5,6, these factors are of limited prognostic utility. Moreover, few studies have integrated sequence data with clinical and cytogentic factors to build predictive models of patient outcome. Here, we sought to identify genomic predictors of refractory disease or early relapse. We used whole genome and exome sequencing to analyze the genomes of 71 adult de novo AML patients treated with anthracycline and cytarabine-based induction chemotherapy. Of these, 34 had refractory disease or relapsed within 6 months, 12 relapsed in 6-12 months, and 25 had a long first remission ( & gt;12 months). We also developed an enhanced exome sequencing (EES) approach to identify and follow leukemia-associated variants over time. In 12 additional patients that achieved morphologic remission after induction chemotherapy, we used EES to identify and track variants at time of diagnosis, time of morphologic remission (roughly 30 days later), and a final time point corresponding to eventual relapse (n=8) or extended remission (n=4). No novel coding or non-coding variants present at the time of diagnosis were found to be predictive of refractory disease or early relapse. Using EES, however, we were able to detect leukemia-associated variants in the initial remission bone marrow in all eight patients who eventually relapsed. One persistent leukemia-associated variant was also detected in one patient still in remission, but all other variants in that patient were eliminated. We also detected 64 somatic variants that became enriched following chemotherapy, but were not detected in the original leukemic cells. These may represent relapse-specific variants or oligoclonal hematopoiesis after bone marrow recovery. Overall, our data suggest that the persistence of leukemia-associated variants after bone marrow recovery from cytotoxic therapy is strongly correlated with relapse, and may be used to complement more traditional, morphologic measures of leukemic cell clearance. 1. Cancer Genome Atlas Research N. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. The New England Journal of Medicine 2013;368:2059-74. 2. Byrd JC, Mrozek K, Dodge RK, et al. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461). Blood 2002;100:4325-36. 3. Grimwade D, Hills RK, Moorman AV, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood 2010;116:354-65. 4. Schlenk RF, Dohner K, Krauter J, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. The New England Journal of Medicine 2008;358:1909-18. 5. Kihara R, Nagata Y, Kiyoi H, et al. Comprehensive analysis of genetic alterations and their prognostic impacts in adult acute myeloid leukemia patients. Leukemia 2014;28:1586-95. 6. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. The New England Journal of Medicine 2010;363:2424-33. This abstract is also presented as a poster at the Translation of the Cancer Genome conference. Citation Format: Jeffery M. Klco, Christopher A. Miller, Malachi Griffith, Allegra Petti, David H. Spencer, Shamika Ketkar-Kulkarni, Lukas D. Wartman, Matthew Christopher, Tamara L. Lamprecht, Jacqueline E. Payton, Jack Baty, Sharon E. Heath, Obi L. Griffith, Dong Shen, Jasreet Hundal, Gue Su Chang, Robert S. Fulton, Michelle O'laughlin, Catrina Fronick, Vincent Magrini, Ryan Demeter, David E. Larson, Shashikant Kulkarni, Bradley A. Ozenberger, John S. Welch, Matthew J. Walker, Timothy A. Graubert, Peter Westervelt, Jerald P. Radich, Daniel C. Link, Elaine R. Mardis, John F. DiPersio, Richard K. Wilson. Genomic approaches for risk assessment in acute myeloid leukemia. [abstract]. In: Proceedings of the AACR Special Conference on Computational and Systems Biology of Cancer; Feb 8-11 2015; San Francisco, CA. Philadelphia (PA): AACR; Cancer Res 2015;75(22 Suppl 2):Abstract nr PR03.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. NG11-NG11
    Abstract: In infectious disease, polyclonal T cell responses against immunodominant epitopes drive successful immune responses. In cancer, neoepitopes (neoE) derived from non-synonymous mutations, similarly to the immunodominant epitopes in viral infections, are potentially highly immunogenic because the T cells recognizing these antigens are not subjected to the mechanisms of tolerance. Indeed, early studies support that neoE derived from non-synonymous mutations are the primary target of T cell responses induced by immune checkpoint blockade therapy and have been successfully targeted by adoptively transferred T cell therapies (ACT) in multiple cancer histologies. However, there is limited knowledge on the immunodominance and evolution of neoE's, or the clonality of the T cell responses against these neoE. Furthermore, little is known regarding the correlation between the presence and expansion of neoE-specific T cells and the clinical response to immunotherapy in patients. To characterize the neoE-specific T cell responses induced after immunotherapy, we collected peripheral blood mononuclear cells (PBMCs) over time (longitudinally) and established expanded tumor infiltrating lymphocyte cultures (TILs) and autologous tumor cell lines from the patient's tumor biopsies. We performed whole exome and RNA sequencing of the tumor and normal tissue controls for the computational prediction and ranking of patient-specific neoEs. We then generated a library of capture reagents consisting of the patient HLA class I molecules loaded with predicted neoE (Peng et al. AACR 2019) and isolated neoE-specific T cells from the patients' PBMC or TIL samples. Once isolated, the paired neoE-specific TCR alpha and beta chains (neoTCR) were obtained by single cell sequencing. For functional characterization of the neoTCRs, healthy donor primary human T cells were modified to express the neoTCR using CRISPR-based, non-viral precision genome engineering by replacing the endogenous TCR with the respective neoTCR (Jacoby et al., AACR 2019, Sennino et al., AACR 2019). These gene-edited T cells were then used in co-culture experiments with the patient autologous cell lines. We analyzed T cell responses in three patients (PT1, PT2, and PT3) with metastatic melanoma receiving immunotherapy. PT1 had a fast and durable anti-tumor response to anti-PD-1 therapy. Sequencing identified 2556 somatic coding mutations. A library of 243 neoE-specific pMHC capture reagents across 3 HLA types, HLA-A*03:01, A*24:01, and C*12:03 was generated and used for screening of PBMCs or TILs derived from multiple longitudinal time points. Several hundred neoE-specific T cells were isolated. Importantly, this neoE-specific T cell response was comprised of 17 different neoE-specific T cells clones targeting only 5 different HLA-neoE complexes supporting the immunodominance hypothesis. On the other hand, PT2 and PT3 showed marginal responses to immunotherapy. Patient two progressed after being treated with anti-PD1. This patient had 24 somatic coding mutations. Seventeen neoE-HLA reagents across 3 HLAs, B*35:03, C*12:03, and C*08:01 were generated and used to capture neoE-specific T cells from TILs and PBMCs. While 14 different TCRs targeting 7 HLA-neoE complexes were identified from expanded TILs, no neoE-reactive T cells were captured from the peripheral blood. PT3 presented with progressive disease after being treated with local TVEC. This patient had 61 somatic coding mutations; 78 neoE-specific pHLA capture reagents covering HLA-A*02:01, A*03:01, B*07:02, C*05:01, and C*07:02 were generated and used to screen for neoE-specific T cells in the patient's TIL and PBMCs. In contrast to PT2, 2 different neoTCRs targeting the same HLA-neoE complexes were isolated from PBMCs, but none from TILs. To further characterize the T cell responses from patients that responded or did not respond to immunotherapy, we generated 18 separate T cell products, each expressing a different neoTCR isolated from PT1, PT2 and PT3. For PT1, we characterized 14 different neoTCRs specific for neoE's in the mutated IL8, PUM1 and TPP2 genes. All 14 T cell products displayed specific cytotoxicity against the matched autologous melanoma cell line established from a biopsy of patient one (50-75% tumor growth inhibition compared to melanoma cell line growth in co-culture with a mismatched control TCR, 96 hour assay using a product to target ratio (P:T) of 1:1, p & lt; 0.000001 for each comparison). No cytotoxic effect against an unmatched human melanoma cell line was observed. Furthermore, neoE TCR T cells upregulated 4-1BB and OX-40, secreted IFNγ, IL-2, TNFα, and IL6, and induced T cell proliferation and degranulation. Again, no unspecific T cell activation was observed when T cells were co-cultured with unmatched targets. Interestingly, precision genome engineered T cell products expressing neoTCRs identified from patients that did not respond to therapy (PT2 and PT3), also potently killed autologous tumor cells. Four neoTCRs were studied (2 TCR for PT2 and 2 TCRs for PT3), and three of them showed specific cytotoxicity against the matched autologous melanoma cell line (50-100% tumor growth inhibition compared to melanoma cell line growth in co-culture with a mismatched control TCR, 96 hour assay using P:T 5:1, p & lt; 0.05 for each comparison). Additionally, upon co-culture with the matched melanoma cell line, but not against an unmatched melanoma cell line control, neoE TCR T cells upregulated 4-1BB and OX-40, secreted IFNγ, IL-2, TNFα, and IL6, and induced T cell proliferation and degranulation. These data demonstrate that even patients that did not respond to immunotherapy harbor neoTCRs that, when expressed in ‘fresh' T cells, are able to kill the autologous tumor cell lines. Using newly developed techniques to isolate and capture neoE-specific single T cells, as well as non-viral gene editing, we isolated and characterized neoE-specific T cells that can recognize the cancer cells and induce an anti-tumor response. We also studied the neoE immunodominance and TCR clonality over time of the natural T cell repertoire that induce anti-tumor responses to ICB therapy. Our results show that in a patient with a good response to anti-PD-1, there is a polyclonal response that targets a limited number of neoE-HLA complexes (2% of the neoE tested in the case of patient one) highlighting the immunodominance of these epitopes. Interestingly, different T cell clonotypes targeting the same mutations evolve over time, suggesting functional differences amongst the TCRs. In addition, our results demonstrate that even patients that did not respond to these therapies harbor neoE-specific T cells, as we were able to isolate neoE-specific T cells that recognized and killed patient-derived cancer cells. This suggests that even in patients that do not respond to immunotherapy, neoE-specific TCRs can be isolated and could be potentially used for personalized ACT. Finally, our results also show how non-viral precision genome engineering can successfully redirect T cells to neoE-expressing tumors, enabling the personalized ACT. Citation Format: Cristina Puig-Saus, Barbara Sennino, Bhamini Purandare, Duo An, Boi Quach, Songming Peng, Huiming Xia, Sidi Zhao, Zheng Pan, Yan Ma, Justin Saco, Sameeha Jilani, Christine Shieh, Katharine Heeringa, Olivier Dalmas, Robert Moot, Diana Nguyen, William Lu, Kyle Jacoby, Andrew Conroy, Jasreet Hundal, Malachi Griffith, Stefanie Mandl, Alex Franzusoff, Antoni Ribas. Landscape analysis of neoepitope-specific T-cell responses to immunotherapy [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr NG11.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 125, No. 4 ( 2015-01-22), p. 619-628
    Abstract: Dnmt3a-null hematopoietic stem cells (HSCs) cannot sustain long-term hematopoiesis. Cooperating c-Kit mutations drive leukemic transformation of Dnmt3a-null HSCs.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    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: Nature Genetics, Springer Science and Business Media LLC, Vol. 51, No. 1 ( 2019-1), p. 175-179
    Type of Medium: Online Resource
    ISSN: 1061-4036 , 1546-1718
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2019
    detail.hit.zdb_id: 1494946-5
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3500-3500
    Abstract: Abstract 3500 Small non-coding RNAs (sncRNAs) have emerged as important regulators of multiple cellular processes, with a growing body of evidence suggesting their dysregulation and possible pathogenic role in many malignancies, including acute myeloid leukemia (AML). Previous high throughput sncRNA transcriptome sequencing studies have focused on the 15–30 nucleotide fraction that primarily consists of microRNAs (miRNAs), thus excluding many larger, potentially important sncRNAs, such as small-nucleolar RNAs (snoRNAs) and piwiRNAs. To address this “sequencing gap”, we extended our transcriptome sequencing to include RNA species of 15–75 nucleotides in length. Here, we report the small RNA transcriptome sequencing of 28 cases of AML. As a control, we also sequenced the small RNA transcriptome in CD34+ progenitors (n = 4) and promyelocytes (n=2) from healthy donors. The most common class of sncRNA detected in all samples was small-nuclear/snoRNAs (∼40% of reads), followed by miRNAs (∼17% of reads), ribosomal/transfer RNAs (15% of reads), unannotated/miscellaneous sncRNAs (∼15% of reads), and piwiRNAs (∼10% of reads). Partek Genomics Suite was used to identify significantly dysregulated sncRNAs using a p-value 〈 0.05, false discovery rate of 5%, and a minimum 2 fold-change from normal CD34+ cells. Compared to normal CD34+ cells, expression of 158 sncRNAs was significantly increased and 29 were significantly decreased in de novo AML samples. The most striking example of dysregulation was observed in acute promyelocytic leukemia (M3 AML). The DLK1-DIO3 locus at 14q32.2 contains 41 snoRNA genes belonging to the SNORD112–114 family, in addition to a large number of miRNAs. Compared with normal CD34+ cells or non-M3 AML, 31 of the 41 snoRNAs in this locus were massively upregulated (6–2,000 fold) in M3 AML samples. Concordantly large increases (10–1,000 fold) of 20 different miRNAs in this locus were also observed in M3 AML. We extended these findings to an independent cohort of M3 and non-M3 AML cases (n = 187) for which RNA transcriptome sequencing was available (RJW and TJL, personal communication, on behalf of the TCGA AML analysis group). Analysis of this data confirmed massive up-regulation of sncRNAs in the DLK1-DIO3 locus that was restricted to M3 AML. We considered the possibility that the increase in sncRNAs in this locus was secondary to a promyelocyte-specific expression pattern in M3 AML cells, but expression of sncRNAs in normal promyelocytes was lower than that observed in normal CD34+ cells, making this hypothesis unlikely. The DLK1-DIO3 locus is one of the best characterized imprinted regions in the human genome. The paternally-derived protein coding genes in this locus (DLK1, RTL1 and DIO3) showed no dysregulation in M3-AML in contrast to the aberrantly expressed, maternally-derived sncRNAs. While the function of the sncRNAs in this region is largely unknown, there is evidence they may contribute to stem cell pluripotency (Stadtfeld et al. Nature 2010, Liu et al. JBC 2010). Since imprinting at this locus is controlled by methylation of several differentially methylated regions (DMRs), we analyzed array-based methylation data from the TCGA cohort to determine if imprinting was disrupted. No difference in methylation status of the DMRs was observed between M3 and non-M3 AML. Moreover, based on expressed germline single nucleotide polymorphisms, mono-allelic expression of sncRNAs in this locus was preserved. Together, these data show that imprinting of the DKL1-DIO3 locus is not disrupted in M3-AML samples, and that dysregulation of the sncRNAs in this region occur through an imprinting-independent mechanism. In summary, extended small RNA transcriptome sequencing is a valuable new tool to analyze malignant cells; it successfully identified massive dysregulation of sncRNAs in the DKL1-DIO3 locus in M3 AML. The contribution of the aberrantly expressed sncRNAs within this locus to the pathogenesis of M3 AML will require additional study. Disclosures: No relevant conflicts of interest to declare.
    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 ...
  • 7
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 37, No. 15_suppl ( 2019-05-20), p. e21064-e21064
    Abstract: e21064 Background: Merkel cell carcinoma (MCC) is an immunogenic neuroendocrine malignancy with promising responses to immune checkpoint blockade (ICB). Although ICB is standard of care in advanced MCC patients, approximately 50% of patients are resistant to ICB. Thus, it is crucial to identify biomarkers predictive of response. Methods: To understand the genomic landscape of MCC, we performed exome capture sequencing on 27 tumor and matched normal samples from 25 patients with MCC treated at Memorial Sloan Kettering. Herein, we report the analysis of 16 paired samples from 14 patients. Nonsynonymous, high-confidence somatic mutations were identified and tumor reads aligned to the Merkel cell polyoma virus (MCPyV) were quantified. Results: Tumor and matched normal samples were sequenced to a median target coverage depth of 53x and 79x reads, respectively. One sample was not analyzed due to inadequate coverage. The MCPyV genome was detected in 12 of 13 patients (92%). The median somatic mutation burden among analyzed samples was 19 nonsynonymous variants per exome (range: 8 - 120). No recurrent driver mutations were identified in any sample. Four samples lacked potential driver mutations and, among the remaining 11 samples, 36 putatively oncogenic mutations were detected in 33 genes (variant allele frequency: 0.08 – 1), including genes involved in the cell cycle checkpoint ( TP53, RB1), DNA damage repair ( ERCC4, FANCA, FANCD2), PI3K-AKT-mTOR ( PIK3CA, PIK3CG), and Notch ( NOTCH1, NOTCH2) pathways. One sample with undetected MCPyV DNA demonstrated loss of heterozygosity of both TP53 and RB1. Four samples contained strand coordinated clusters of mutations in more than 20 distinct gene regions, suggesting an APOBEC-high mutagenesis signature. Conclusions: The MCPyV genome was detected in most tumors analyzed and tumor mutation burden was low in such tumors, consistent with published literature. Analysis of an additional 11 sample pairs is ongoing, along with personalized neoantigen binding predictions on all samples. Immunohistochemistry (IHC) for PDL1 and CD8 expression is in progress. Associations between the aforementioned and response to ICB will be reported.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2019
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 837-837
    Abstract: Genome sequencing studies of patient samples have implicated the involvement of various components of the epigenetic machinery in myeloid diseases, including the de novo DNA methyltransferase DNMT3A (Cancer Genome Atlas Research, N Engl J Med, 2013). We have recently shown that Dnmt3a is essential for normal hematopoietic stem cell (HSC) differentiation. Genetic ablation of Dnmt3a resulted in HSCs that showed diminished capacity for peripheral blood generation after serial transplantation (on a per-HSC basis), while phenotypically-defined HSCs accumulated in the bone marrow (Challen et al., Nature Genetics, 2012). Although this differentiation arrest was insufficient to cause overt disease, in these competitive transplants the presence of wild-type whole bone marrow may have suppressed malignant transformation of the mutant HSCs. Dnmt3a-null HSCs were less proliferative than counterpart control HSCs in this transplantation setting, suggesting that the cellular turnover threshold necessary to generate additional genetic and/or epigenetic lesions required for leukemogenesis was not achieved. To further understand the contribution of Dnmt3a loss-of-function in hematopoiesis, we performed non-competitive transplantation of Dnmt3a-null bone marrow. This forces the mutant HSCs to divide in vivoto regenerate the hematopoietic system following lethal irradiation, and should uncover any predispositions to transformation. Mice transplanted with Dnmt3a-null bone marrow in the absence of wild-type support cells succumbed principally to bone marrow failure (median survival 328 days) characteristic of myelodysplastic syndromes (MDS) with symptoms including anemia, neutropenia, bone marrow hypercellularity and splenomegaly with myeloid infiltration. 2/25 mice developed myeloid leukemia with 〉 20% blasts in the blood and bone marrow. 4/25 primary mice succumbed to myeloproliferative disorders, some of which progressed to secondary leukemia after long latency. Exome sequencing was performed to identify co-operating mutations that drove leukemic transformation, and revealed c-Kit mutations found only in the Dnmt3a-null AML samples. As DNMT3A and KIT mutations can co-occur in AML and mastocytosis, we tested whether these two pathways could co-operate in vivo by ectopic introduction of c-Kit variants into hematopoietic progenitors followed by bone marrow transplantation (Figure 1). As previously reported, expression of c-KitD814V in wild-type cells lead to development of B-cell acute lymphoblastic leukemia (B-ALL). However, expression of c-KitD814V in a Dnmt3a-null background lead to acute leukemia with a much shorter latency (median survival 67 days), implicating a synergism between these pathways in vivo. Moreover, the absence of Dnmt3a also distorted the spectrum of leukemia resulting from enforced c-Kit signaling. While some of the mice transplanted with Dnmt3a-null c-KitD814V cells also succumbed to a B-ALL, 4/13 (31%) developed mastocytosis with involvement of myeloid blasts, and 4/13 (31%) mice developed a T-cell acute lymphoblastic leukemia (T-ALL). We show for the first time that these pathways can co-operate to accelerate transformation in vivo. This Dnmt3a/c-Kit disease model resembles the classical “two-hit” model of leukemogenesis in which one mutation in a hematopoietic progenitor cell inhibits differentiation (Dnmt3a loss-of-function), whilst another drives proliferation (c-Kit gain-of-function). Such mouse models present a unique opportunity to study the sequence of early events leading to HSC transformation following Dnmt3a-inactivation. Figure 1 Kaplan-Meier survival curve of mice transplanted with control or Dnmt3a-KO bone marrow progenitor cells transduced with a lentivirus expressing c-KitD814V. *** p 〈 0.001. Figure 1. Kaplan-Meier survival curve of mice transplanted with control or Dnmt3a-KO bone marrow progenitor cells transduced with a lentivirus expressing c-KitD814V. *** p 〈 0.001. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    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 ...
  • 9
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 9, No. 1 ( 2018-09-04)
    Abstract: Here we report targeted sequencing of 83 genes using DNA from primary breast cancer samples from 625 postmenopausal (UBC-TAM series) and 328 premenopausal (MA12 trial) hormone receptor-positive (HR+) patients to determine interactions between somatic mutation and prognosis. Independent validation of prognostic interactions was achieved using data from the METABRIC study. Previously established associations between MAP3K1 and PIK3CA mutations with luminal A status/favorable prognosis and TP53 mutations with Luminal B/non-luminal tumors/poor prognosis were observed, validating the methodological approach. In UBC-TAM, NF1 frame-shift nonsense (FS/NS) mutations were also a poor outcome driver that was validated in METABRIC. For MA12, poor outcome associated with PIK3R1 mutation was also reproducible. DDR1 mutations were strongly associated with poor prognosis in UBC-TAM despite stringent false discovery correction ( q  = 0.0003). In conclusion, uncommon recurrent somatic mutations should be further explored to create a more complete explanation of the highly variable outcomes that typifies ER+ breast cancer.
    Type of Medium: Online Resource
    ISSN: 2041-1723
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2553671-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 9, No. 1 ( 2018-11-14)
    Abstract: The original version of this Article contained errors in the depiction of confidence intervals in the NF1 BCSS data illustrated in Figure 3b. These have now been corrected in both the PDF and HTML versions of the Article. The incorrect version of Figure 3b is presented in the associated Author Correction.
    Type of Medium: Online Resource
    ISSN: 2041-1723
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2553671-0
    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