Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2017
    In:  Scientific Reports Vol. 7, No. 1 ( 2017-05-08)
    In: Scientific Reports, Springer Science and Business Media LLC, Vol. 7, No. 1 ( 2017-05-08)
    Abstract: Oncogenic transformation leads to dysregulated cell proliferation, nutrient deficiency, and hypoxia resulting in metabolic stress and increased DNA damage. In normal cells, such metabolic stress leads to inhibition of signaling through the mammalian Target of Rapamycin Complex 1 (mTORC1), reduction of protein translation, cell cycle arrest, and conservation of energy. In contrast, negative regulation of mTORC1 signaling by DNA damage is abrogated in many cancer cells, thus mTORC1 signaling remains active under microenvironmental conditions that potentially promote endogenous DNA damage. Here we report that mTORC1 signaling suppresses endogenous DNA damage and replication stress. Pharmacological inhibition of mTOR signaling resulted in phosphorylation of H2AX concomitant with the decrease of CHK1 levels both in cell culture and mouse rhadomyosarcoma xenografts. Further results demonstrated that mTORC1-S6K1 signaling controls transcription of CHK1 via Rb-E2F by upregulating cyclin D and E. Consistent with these results, downregulation of CHK1 by inhibition of mTOR kinase resulted in defects in the slow S phase progression following DNA damage. These results indicate that, under stressful conditions, maintained mTORC1 signaling in cancer cells promotes survival by suppressing endogenous DNA damage, and may control cell fate through the regulation of CHK1.
    Type of Medium: Online Resource
    ISSN: 2045-2322
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2017
    detail.hit.zdb_id: 2615211-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Impact Journals, LLC ; 2017
    In:  Oncotarget Vol. 8, No. 25 ( 2017-06-20), p. 41422-41431
    In: Oncotarget, Impact Journals, LLC, Vol. 8, No. 25 ( 2017-06-20), p. 41422-41431
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2017
    detail.hit.zdb_id: 2560162-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 22, No. 24 ( 2016-12-15), p. 6142-6152
    Abstract: Purpose: Selinexor, a selective inhibitor of XPO1, is currently being tested as single agent in clinical trials in acute myeloid leukemia (AML). However, considering the molecular complexity of AML, it is unlikely that AML can be cured with monotherapy. Therefore, we asked whether adding already established effective drugs such as topoisomerase (Topo) II inhibitors to selinexor will enhance its anti-leukemic effects in AML. Experimental Design: The efficacy of combinatorial drug treatment using Topo II inhibitors (idarubicin, daunorubicin, mitoxantrone, etoposide) and selinexor was evaluated in established cellular and animal models of AML. Results: Concomitant treatment with selinexor and Topo II inhibitors resulted in therapeutic synergy in AML cell lines and patient samples. Using a xenograft MV4-11 AML mouse model, we show that treatment with selinexor and idarubicin significantly prolongs survival of leukemic mice compared with each single therapy. Conclusions: Aberrant nuclear export and cytoplasmic localization of Topo IIα has been identified as one of the mechanisms leading to drug resistance in cancer. Here, we show that in a subset of patients with AML that express cytoplasmic Topo IIα, selinexor treatment results in nuclear retention of Topo IIα protein, resulting in increased sensitivity to idarubicin. Selinexor treatment of AML cells resulted in a c-MYC–dependent reduction of DNA damage repair genes (Rad51 and Chk1) mRNA and protein expression and subsequent inhibition of homologous recombination repair and increased sensitivity to Topo II inhibitors. The preclinical data reported here support further clinical studies using selinexor and Topo II inhibitors in combination to treat AML. Clin Cancer Res; 22(24); 6142–52. ©2016 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 26, No. 3 ( 2020-02-01), p. 669-678
    Abstract: EGF-like domain 7 (EGFL7) is a secreted protein and recently has been shown to play an important role in acute myeloid leukemia (AML); however, the underlying mechanism by which EGFL7 promotes leukemogenesis is largely unknown. Experimental Design: Using an antibody interaction array, we measured the ability of EGFL7 to bind directly approximately 400 proteins expressed by primary AML blasts. Primary patient samples were stimulated in vitro with recombinant EGFL7 (rEGFL7) or anti-EGFL7 blocking antibody to assess alterations in downstream signaling and the ability to effect blast differentiation and survival. We treated three independent AML models with anti-EGFL7 or IgG1 control to determine whether anti-EGFL7 could prolong survival in vivo. Results: We found EGFL7 significantly binds several signaling proteins important for normal and malignant hematopoiesis including NOTCH. Stimulation of AML blasts with rEGFL7 reduced NOTCH intracellular domain and NOTCH target gene expression while treatment with an anti-EGFL7 blocking antibody resulted in reactivation of NOTCH signaling, increased differentiation, and apoptosis. Competitive ligand-binding assays showed rEGFL7 inhibits DELTA-like (DLL) 4-mediated NOTCH activation while anti-EGFL7 combined with DLL4 significantly increased NOTCH activation and induced apoptosis. Using three different AML mouse models, we demonstrated that in vivo treatment with anti-EGFL7 alone results in increased survival. Conclusions: Our data demonstrate that EGFL7 contributes to NOTCH silencing in AML by antagonizing canonical NOTCH ligand binding. Reactivation of NOTCH signaling in vivo using anti-EGFL7 results in prolonged survival of leukemic mice, supporting the use of EGFL7 as a novel therapeutic target in AML.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 519-519
    Abstract: Background: A HOXB-locus-embedded lncRNA, named HOXB-AS3 significantly associates with NPM1 mutations in AML. Herein, we evaluate the functional role of HOXB-AS3 expression in NPM1mut AML. Methods: HOXB-AS3 expression was measured by real-time PCR. Knock-down (KD) of HOXB-AS3 was performed in vitro and in vivo with locked nucleic acid-modified gapmers. RNA antisense purification (RAP), RNA-immunoprecipitation (RIP), and Chromatin-immunoprecipitation (ChIP) experiments were performed according to published protocols. Results: Among 7 AML cell lines tested, only OCI-AML3 cells, which harbor NPM1mut, showed detectable HOXB-AS3 expression. HOXB-AS3 was more abundant in NPM1mut AML patient (pt) blasts than blasts of AML pts with wild-type NPM1 (P=.001) and bone marrow samples from healthy donors(P=.001). HOXB-AS3 localized in the nucleus and did not associate with isolated polysomes of OCI-AML3 cells. In vitro HOXB-AS3 KD in OCI-AML3 cells decreased the cells in S phase (P & lt;.001) and increased those in G2/M phase (P=.006). HOXB-AS3 KD reduced the number of formed colonies by OCI-AML3 cells (P=.02). In contrast, overexpression of HOXB-AS3 in K562 cells increased the cells in S phase (P=.02) and decreased those in G0/G1 phase (P=.008). HOXB-AS3 KD in blasts of 3 NPM1mut AML pts decreased the number of formed colonies (P=.03, P=.02, and P & lt;.001). In vivo HOXB-AS3 KD in murine patient-derived xenografts of 2 NPM1mut AML pts prolonged their overall survival (P & lt;.001 and P=.03). RAP-based isolation of HOXB-AS3 and comparative proteomic analyses identified 23 candidate HOXB-AS3-binding proteins. EBP1 was validated as the most avid HOXB-AS3 interactor (P & lt;.001) by RIP experiments. Manipulations of HOXB-AS3 impacted on the (previously reported) EBP1 interaction with NPM1; HOXB-AS3 KD reduced, whereas overexpression of HOXB-AS3 increased the EBP1-NPM1 complex formation. Consequently, HOXB-AS3 KD reduced transcription of rRNA and de novo protein synthesis in OCI-AML3 cells (P & lt;.001 and P=.002) and AML pt blasts (P & lt;.001 and P=.03, respectively). Overexpression of HOXB-AS3 increased rRNA transcription (P & lt;.001), de novo protein synthesis (P=.001). ribosomal DNA (rDNA) promoter occupancy by RNA-Polymerase I (P=.001), and activity of an rDNA promoter-containing luciferase reporter (P=.002) in K562 cells. We hypothesized that HOXB-AS3 guides EBP1 to the rDNA locus. RAP-DNA experiments validated the interaction of HOXB-AS3 with rDNA chromatin (P=.001) and HOXB-AS3 KD decreased the occupancy of the rDNA promoter by EBP1 (P=.002), as shown by ChIP assays. Conclusions: We describe the function of the HOXB-AS3 lncRNA as a compensatory mechanism, which mediates increased rRNA transcription and adequate protein production, in NPM1mut AML. From a therapeutic standpoint, we show that HOXB-AS3-targeting yields anti-leukemic activity in pre-clinical models. Citation Format: Dimitrios Papaioannou, Andreas Petri, Sara Terreri, Charlotte A. Thrue, Deedra Nicolet, Frances A. Collins, Lauren A. Woodward, Prasanthi Kumchala, Malith Karunasiri, Felice Pepe, Marius Bill, Nina Zitzer, Guramrit Singh, Sakari Kaupinnen, Clara D. Bloomfield, Adrienne M. Dorrance, Ramiro Garzon. The long non-coding RNA (lncRNA) HOXB-AS3 regulates transcription of ribosomal RNA (rRNA) in NPM1-mutated (NPM1mut) acute myeloid leukemia (AML) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 519.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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 ...
  • 6
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 184, No. 6 ( 2010-03-15), p. 2769-2775
    Abstract: IL-15 is required for NK cell development and homeostasis in vivo. Because IL-15 is presented in trans via its high-affinity IL-15Rα–chain to cells expressing the IL-15Rβγ complex, we postulated that certain IL-15–bearing cells must be required for NK cell homeostasis. Using IL-15WT/WT and IL-15−/− mice, bone marrow chimeras with normal cellularity, and a selective depletion of CD11chi dendritic cells (DCs), we demonstrate that ablation of the resting CD11chi DC population results in a highly significant decrease in the absolute number of mature NK cells. In contrast, administration of Flt3 ligand increases the CD11chi DC population, which, when expressing IL-15, significantly expands mature NK cells via enhanced survival and proliferation. In summary, a CD11chi DC population expressing IL-15 is required to maintain NK cell homeostasis under conditions of normal cellularity and also is required to mediate Flt3 ligand-induced NK cell expansion in vivo.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2010
    detail.hit.zdb_id: 1475085-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Experimental Hematology, Elsevier BV, Vol. 111 ( 2022), p. S149-S150
    Type of Medium: Online Resource
    ISSN: 0301-472X
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    detail.hit.zdb_id: 2005403-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 150-150
    Abstract: Abstract 150 Background. The Mll PTD and Flt3 ITD are co-present in a subset of adult patients (pts) with cytogenetically normal (CN) acute myeloid leukemia (AML) and poor clinical outcomes. While the single mutant knock-in (KI) mice (Mll PTD or Flt3 ITD) exhibit enhanced myeloid progenitor self-renewal or reduced apoptosis, respectively, neither model develops acute leukemia. We hypothesized that with mutant expression driven via the endogenous promoters, the two mutations may cooperate in vivo to induce an acute leukemia that mimics the human counterpart. Methods. Single mutant heterozygous KI mice were crossed to produce the PTD/ITD double KI. PTD/ITD mice were bred with the homozygous Flt3 ITD to generate the PTD/ITD2 genotype. An AML diagnosis was based on blood differentials, immunophenotyping, tissue pathology and transplantability. Real time RT-PCR and 5'-methylcytosine LC/MS assays measured gene expression and global DNA methylation levels, respectively. Results. PTD/ITD and PTD/ITD2 mice developed transplantable, CN-AML/undifferentiated leukemia exhibiting expansion of monocytic/myelomonocytic Gr1±/Mac1+ and/or immature CD3−/CD19−/CD117+/Mac1−/B220lo cell populations, splenomegaly, leukocytosis, anemia and thrombocytopenia. PTD/ITD mice had significantly reduced lifespans compared to mice with single mutant PTD and ITD KIs and wild-type (Wt) controls (medians: 50, 99, 88, 94 weeks, respectively; P 〈 0.001) (Figure 1). Increased ITD gene dosage (PTD/ITD2) was associated with an even shorter lifespan (median: 16 weeks) (Figure 1). This is consistent with the poor prognosis conferred by high FLT3 ITD-to-FLT3 wild-type (WT) gene ratio in diagnostic leukemia blasts from AML pts treated with intensive chemotherapy. As in human MLL PTD AML, the Mll WT allele was downregulated in the murine model. Mll WT expression was 〉 2-fold lower in bone marrow (BM) of leukemic PTD/ITD mice compared to age-matched single mutant KIs or Wt controls. HoxA9 and its cofactor Meis1 were upregulated 15- and 5-fold, respectively, in PTD/ITD mice with leukemia versus Wt BM. Yet, compared to Wt BM, single PTD KI exhibited increased HoxA9 (∼6-fold) but not Meis1, implicating an expression threshold for HoxA9 and a crucial role for Meis1 for the development of acute leukemia in the double KI. Consistent with Flt3 being a downstream transcriptional target of Meis1, total Flt3 mRNA (WT and ITD) levels increased 3-fold in the leukemic PTD/ITD mice relative to either single mutant KIs or Wt controls. Furthermore, one consequence of constitutive Flt3 ITD kinase activity is the upregulation of the anti-apoptotic kinase, Pim1, in human AML. Compared to Wt BM, a 2-fold increase in Pim-1 expression was observed in single ITD KI and a 6-fold increase was observed in leukemic PTD/ITD BM, while expression was unchanged in the single PTD KI BM. Finally, MLL PTD presence in human AML associates with increased global DNA methylation and silencing of tumor suppressor genes. We observed 3-fold higher transcript levels of a de novo methyltransferase, DNA methyltransferase 3b (DNMT3b), increased global DNA methylation and ≥2-fold decrease in the expression of tumor suppressors Id4, Shp1 and Cdkn1b in BM of leukemia PTD/ITD mice compared to age-matched single mutant KIs and Wt controls. Conclusion. The Flt3 ITD and Mll PTD, expressed via their endogenous promoters, cooperate in vivo to give rise to AML and acute undifferentiated leukemia. Elevations of Meis1 and DNMT3b solely in PTD/ITD animals appear to be critical points of dysregulation leading to development of acute leukemia. This novel murine model phenotypically, molecularly, and epigenetically mimics the human AML counterpart, thus making it highly relevant for examining critical pathways in acute myeloid leukemogenesis, investigating leukemia stem/initiating cell biology and microenvironment contributions, and testing novel targeting therapeutics. 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: 2010
    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: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2736-2736
    Abstract: Abstract 2736 BCR-ABL1 –positive chronic myeloid leukemia in chronic phase (CML-CP) is a leukemia stem cell (LSC)-derived but leukemia progenitor cell (LPC)-driven disease, which may eventually develop resistance to the tyrosine kinase inhibitors (TKIs) and progress to fatal CML blast phase (CML-BP). In CML-CP, LSCs and LPCs reside in the CD34+CD38- and CD34+CD38+ populations, respectively. In addition, majority of LSCs and LPCs belong to quiescent (CFSEmax) and proliferative (CFSElow) populations, respectively. Quiescent LSCs are intrinsically insensitive to TKIs, and LPCs can acquire resistance to TKIs. In the TKI era, these cells may eventually initiate the disease relapse and progression to CML-BP, which is associated with genomic instability manifested by accumulation of a new or additional TKI-resistant BCR-ABL1 kinase mutations and chromosomal aberrations. We reported that BCR-ABL1 –positive leukemia cells contain high levels of the reactive oxygen species (ROS)-induced oxidative DNA damage resulting in genomic instability (Nowicki et al., Blood, 2005; Koptyra et al., Blood, 2006; Koptyra et al., Leukemia, 2008). These studies highlighted the importance of identification of the origin of leukemia cell lineage accumulating genomic instability and the mechanisms responsible for generation of ROS-mediated oxidative DNA damage. Here we show that LSC-enriched CD34+CD38- cells and quiescent LSCs, and also LPC-enriched CD34+CD38+ and proliferating CML-CP cells contain higher levels of ROS (superoxide anion, hydrogen peroxide, and hydroxyl radical) and oxidative DNA lesions (8-oxoG and DNA double-strand breaks) than corresponding cells from healthy donors. Surprisingly, the most primitive quiescent LSCs accumulated the highest levels of ROS and oxidative DNA damage. On the basis of these observations and the studies in murine hematopoietic 32Dcl3 cells expressing TKI-resistant BCR-ABL1 kinase variants (Y253F, T315I, H396P), we would predict that primitive CML-CP cells carrying these mutations would also contain high levels of ROS and oxidative DNA damage. Moreover, inhibition of BCR-ABL1 kinase with imatinib exerted only modest, if any, effect on ROS and oxidative DNA damage in LSCs/LPCs in the presence of growth factors (GFs). Among numerous signaling proteins activated in CML cells, Rac GTPases were potential candidates to regulate production of ROS. Importantly, Rac was stimulated in leukemia cells expressing non-mutated BCR-ABL1 and TKI-resistant kinase mutants and it remained active in CML-CP cells treated with imatinib in the presence of GFs. We used Rac dominant-negative mutant (RacT17N), Rac specific inhibitors (NSC23766 and EHT1864) and Rac1, Rac2 and Rac3 knockout cells to document that Rac2 GTPase is responsible for elevation of ROS and oxidative DNA damage in LSC-enriched CD34+CD38- cells, quiescent LSCs, and also in LPCs. Active Rac2 reduced mitochondrial membrane potential (ΔΨm) and slowed the electron flow between mitochondrial respiratory chain (MRC) complexes I-II and I-III leading to overproduction of ROS. Using cells depleted of functional mitochondria (Rho0 cells), applying specific probes to measure mitochondrial ROS (MitosoxRed and mitochondria matrix-targeted circularly permuted yellow fluorescence protein = mt-cpYFP) and employing a specific inhibitor of mitochondrial ROS (MitoQ) we determined that mitochondria are the main source of ROS causing oxidative DNA damage in CD34+CD38- and quiescent LSCs and in LPCs. Furthermore, using selective inhibitors of various MRC complexes we pinpointed complex III as major producer of ROS in LSCs and LPCs. This conclusion is supported by the observation that BCR-ABL1 –positive cells with genetically inactivated complex III, but not complex I, displayed diminished capability to generate ROS. Targeting Rac2 GTPase by RacT17N and reduction of mitochondrial ROS by mitochondrial-targeted catalase and by mitochondrial-targeted ROS-scavenging peptide aptamers prevented genomic instability. Altogether, Rac2 - MRC-cIII pathway is a major source of ROS-mediated oxidative DNA damage resulting in genomic instability in LSCs and LPCs, which could be targeted to prevent the relapse and malignant progression of CML. We also postulate that similar mechanisms cause genomic instability in FLT3(ITD)-positive acute myeloid leukemia cells and in JAK2(V617F)-positive polycythemia vera cells. Disclosures: Holyoake: Novartis: Consultancy, Research Funding. Valent:Novartis: Consultancy, Honoraria, Research Funding. Hochhaus:Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Ariad: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. Hughes:BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Ariad: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    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 ...
  • 10
    Online Resource
    Online Resource
    MDPI AG ; 2022
    In:  International Journal of Molecular Sciences Vol. 23, No. 20 ( 2022-10-14), p. 12271-
    In: International Journal of Molecular Sciences, MDPI AG, Vol. 23, No. 20 ( 2022-10-14), p. 12271-
    Abstract: Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm initiated by the presence of the fusion gene BCR::ABL1. The development of tyrosine kinase inhibitors (TKIs) highly specific to p210BCR-ABL1, the constitutively active tyrosine kinase encoded by BCR::ABL1, has greatly improved the prognosis for CML patients. Now, the survival rate of CML nearly parallels that of age matched controls. However, therapy resistance remains a persistent problem in the pursuit of a cure. TKI resistance can be attributed to both BCR::ABL1 dependent and independent mechanisms. Recently, the role of non-coding RNAs (ncRNAs) has been increasingly explored due to their frequent dysregulation in a variety of malignancies. Specifically, microRNAs (miRNAs), circular RNAs (circRNAs), and long non-coding RNAs (lncRNAs) have been shown to contribute to the development and progression of therapy resistance in CML. Since each ncRNA exhibits multiple functions and is capable of controlling gene expression, they exert their effect on CML resistance through a diverse set of mechanisms and pathways. In most cases ncRNAs with tumor suppressing functions are silenced in CML, while those with oncogenic properties are overexpressed. Here, we discuss the relevance of many aberrantly expressed ncRNAs and their effect on therapy resistance in CML.
    Type of Medium: Online Resource
    ISSN: 1422-0067
    Language: English
    Publisher: MDPI AG
    Publication Date: 2022
    detail.hit.zdb_id: 2019364-6
    SSG: 12
    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