Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 117, No. 12 ( 2011-03-24), p. 3409-3420
    Abstract: In BCR-ABL1+ leukemia, drug resistance is often associated with up-regulation of BCR-ABL1 or multidrug transporters as well as BCR-ABL1 mutations. Here we show that the expression level of the transcription factor STAT5 is another parameter that determines the sensitivity of BCR-ABL1+ cells against tyrosine kinase inhibitors (TKIs), such as imatinib, nilotinib, or dasatinib. Abelson-transformed cells, expressing high levels of STAT5, were found to be significantly less sensitive to TKI-induced apoptosis in vitro and in vivo but not to other cytotoxic drugs, such as hydroxyurea, interferon-β, or Aca-dC. The STAT5-mediated protection requires tyrosine phosphorylation of STAT5 independent of JAK2 and transcriptional activity. In support of this concept, under imatinib treatment and with disease progression, STAT5 mRNA and protein levels increased in patients with Ph+ chronic myeloid leukemia. Based on our data, we propose a model in which disease progression in BCR-ABL1+ leukemia leads to up-regulated STAT5 expression. This may be in part the result of clonal selection of cells with high STAT5 levels. STAT5 then accounts for the resistance against TKIs, thereby explaining the dose escalation frequently required in patients reaching accelerated phase. It also suggests that STAT5 may serve as an attractive target to overcome imatinib resistance in BCR-ABL1+ leukemia.
    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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 119, No. 18 ( 2012-05-03), p. 4242-4252
    Abstract: Aggressive systemic mastocytosis (ASM) and mast cell leukemia (MCL) are advanced hematopoietic neoplasms with poor prognosis. In these patients, neoplastic mast cells (MCs) are resistant against various drugs. We examined the effects of 2 demethylating agents, 5-azacytidine and decitabine on growth and survival of neoplastic MCs and the MC line HMC-1. Two HMC-1 subclones were used, HMC-1.1 lacking KIT D816V and HMC-1.2 exhibiting KIT D816V. Both agents induced apoptosis in HMC-1.1 and HMC-1.2 cells. Decitabine, but not 5-azacytidine, also produced a G2/M cell-cycle arrest in HMC-1 cells. Drug-induced apoptosis was accompanied by cleavage of caspase-8 and caspase-3 as well as FAS-demethylation and FAS–re-expression in neoplastic MCs. Furthermore, both demethylating agents were found to synergize with the FAS-ligand in inducing apoptosis in neoplastic MCs. Correspondingly, siRNA against FAS was found to block drug-induced expression of FAS and drug-induced apoptosis in HMC-1 cells. Neither 5-azacytidine nor decitabine induced substantial apoptosis or growth arrest in normal MCs or normal bone marrow cells. Together, 5-azacytidine and decitabine exert growth-inhibitory and proapoptotic effects in neoplastic MCs. These effects are mediated through “FAS–re-expression” and are augmented by the FAS-ligand. Whether epigenetic drugs produce antineoplastic effects in vivo in patients with ASM and MCL remains to be determined.
    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. 118, No. 21 ( 2011-11-18), p. 3457-3457
    Abstract: Abstract 3457 Aggressive systemic mastocytosis (ASM) and mast cell leukemia (MCL) are advanced myeloid neoplasms with a poor prognosis. In these patients, neoplastic mast cells (MC) are resistant against most conventional drugs. Demethylating agents reportedly exert beneficial effects in several advanced myelogenous neoplasms, including myelodysplastic syndromes. We examined the effects of two demethylating agents, 5-Azacytidine and 5-Aza-2`Deoxycytidine (Decitabine) on growth and survival (apoptosis) of neoplastic MC and the human MC line HMC-1. Two HMC-1 subclones were used, HMC-1.1 lacking KIT D816V and HMC-1.2 exhibiting KIT D816V. Both demethylating agents were found to induce apoptosis and growth inhibition in HMC-1.1 cells and HMC-1.2 cells in a dose-dependent manner (IC50: 5-Azacytidine: 5–10 μM, Decitabine: 1–5 μM). Interestingly, only Decitabine but not 5-Azacytidine induced a major G2/M cell cycle arrest in HMC-1 cells. Drug-induced apoptosis in HMC-1 cells was accompanied by cleavage and activation of Caspase-8 and Caspase-3 as well as an increased expression of proapoptotic FAS/CD95, whereas no major effects on expression of other surface antigens were seen. We also found that both demethylating agents synergize with the FAS-ligand in inducing apoptosis in neoplastic MC. Methylation-specific PCR and bisulfite genomic sequencing revealed that the FAS-promoter is hypermethylated in HMC-1 cells. In addition, qPCR demonstrated that exposure to 5-Azacytidine or Decitabine leads to re-expression of FAS in neoplastic MC, which was confirmed by flow cytometry. Correspondingly, a FAS-specific siRNA was found to block drug-induced expression of FAS and drug-induced apoptosis in HMC-1 cells. Although other key regulators and tumor suppressor molecules such as p16 were also found to be hypermethylated in HMC-1 cells, no major demethylating effects of 5-Azacytidine or Decitabine were seen. Neither 5-Azacytidine nor Decitabine induced substantial apoptosis or growth arrest in normal human cord blood progenitor-derived MC or in control bone marrow cells. Together, our data show that 5-Azacytidine and Decitabine exert growth-inhibitory and pro-apoptotic effects in neoplastic MC. These effects are mediated through FAS re-expression and are augmented by the FAS ligand. Whether epigenetic drugs produce anti-neoplastic effects in vivo in patients with advanced SM including MCL, remains to be determined in clinical trials. Disclosures: Valent: Novartis: Consultancy, Honoraria, 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 ...
  • 4
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1654-1654
    Abstract: Acute lymphoblastic leukemia (ALL) is a life-threatening hematopoietic neoplasm characterized by abnormal growth and accumulation of lymphatic blast cells in various hematopoietic tissues. In a substantial number of patients, the Philadelphia (Ph) chromosome and the related oncoprotein BCR/ABL, are detectable. Despite recent advances in the management and therapy of patients with ALL, including the use of BCR/ABL1 tyrosine kinase inhibitors (TKI), the prognosis is still poor. Therefore, several attempts have been made to improve targeted treatment approaches in ALL. One strategy is to identify markers and targets expressed on leukemic stem cells (LSC) in these patients and to apply targeted drugs in order to eliminate LSC. In patients with Ph+ ALL, the leukemia-initiating cell-population is considered to reside within a CD34+/CD38- fraction of the clone. In the present study, we examined the expression of various stem cell markers and target antigens in CD34+/CD38- stem cells and in more mature CD34+/CD38+ progenitor cells in patients with Ph+ ALL (n=12), Ph- ALL (n=13), Ph+ CML (n=20), and in control bone marrow (BM) samples (unexplained cytopenia, n=10). Surface expression of target antigens was analyzed by multicolor flow cytometry, and mRNA expression levels by qPCR. As assessed by flow cytometry, CD34+/CD38- cells were found to co-express CD19, the stem cell-homing receptor CD44, the Campath-1 antigen (CD52), AC133 (CD133), FLT3 (CD135), and CXCR4 (CD184) in all ALL patients examined. In a majority of the ALL patients tested (14/25), LSC also expressed Siglec-3 (CD33). In CML, LSC were found to express a similar profile of antigens, including CD33, CD44, CD52, CD133, CD135, and CXCR4, but these cells did not express CD19. In control BM samples, CD34+/CD38- cells expressed a similar phenotype, but the levels of CD33 and CD52 were lower compared to LSC in ALL and CML. The IL-1RAP was found to be expressed on LSC in patients with Ph+ CML and Ph+ ALL, but not on LSC in Ph- ALL or in normal BM stem cells. By contrast, the SCF receptor KIT (CD117) was found to be expressed on LSC in Ph+ CML but was hardly detectable on LSC in patients with Ph+ ALL or Ph- ALL. The IL-2RA (CD25) and the SDF-1-degrading surface enzyme dipeptidyl-peptidase IV (DPPIV=CD26) were expressed on LSC in patients with CML and in all patients with Ph+ ALL exhibiting BCR/ABL-p210, whereas in Ph+ ALL with BCR/ABL-p190, LSC variably expressed CD25, and did not express CD26. In patients with Ph- ALL and in the normal BM, CD34+/CD38- cells did not express CD25 or CD26. The target receptor CD20 was detectable on ALL LSC in 7/18 patients examined. All target receptors tested were also detectable on more mature CD34+/CD38+ progenitor cells in patients with Ph+ ALL and Ph- ALL. In consecutive studies, expression of target antigens was confirmed at the mRNA level by qPCR analyses of highly enriched ALL LSC. Finally, we were able to show that the CD52-targeting drug alemtuzumab induces rapid lysis of CD34+/CD38- ALL LSC in all patients examined (Figure). In summary, our data show that LSC in Ph+ ALL and Ph- ALL express a unique phenotype, including clinically relevant cytokine receptors and cell surface target antigens, including the Campath-1 antigen, CD52. In Ph+ ALL with BCR/ABL-p210, the phenotype of ALL LSC largely resembles the phenotype of LSC in Ph+ CML, confirming the close relationship and similar pathogenesis of these two types of leukemias. Ficoll-isolated MNC of 4 patients with Ph+ ALL were incubated in control medium (Co) or in various concentrations of alemtuzumab (10-300 µg/ml) in RPMI 1640 medium in the presence of 30% human serum at 37°C for 1 hour. After washing, cells were stained with fluorochrome-conjugated mAb against CD34, CD38 and CD45 for 15 minutes. DAPI-staining was used to evaluate the percentage of viable cells. Cells were analysed using a FACSCanto II and FlowJo software. Results show the numbers of viable CD34+/CD38- cells and are expressed as percent of control (Co). Values represent the mean±S.D. of four independent experiments. Asterisk (*): p 〈 0.05 compared to control. Disclosures: Valent: Novartis: Consultancy, 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 ...
  • 5
    In: The FASEB Journal, Wiley, Vol. 28, No. 8 ( 2014-08), p. 3540-3551
    Type of Medium: Online Resource
    ISSN: 0892-6638 , 1530-6860
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2014
    detail.hit.zdb_id: 1468876-1
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: The FASEB Journal, Wiley, Vol. 26, No. 2 ( 2012-02), p. 894-906
    Type of Medium: Online Resource
    ISSN: 0892-6638 , 1530-6860
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2012
    detail.hit.zdb_id: 1468876-1
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 961-961
    Abstract: Abstract 961 In Philadelphia-positive (Ph+) chronic myeloid leukemia (CML), leukemic stem cells (LSC) supposedly reside in a CD34+/CD38−/Lin− fraction of the leukemic clone. However, little is known about phenotypic properties of LSC in CML. We screened for novel LSC markers and targets in CML by gene chip studies and extensive flow cytometry analyses using monoclonal antibodies against various surface antigens (n=50). A total number of 240 bone marrow or peripheral blood samples (CML, n=95; AML, n=103; CMML, n=10, control marrow, n=32) were examined. In common with normal SC, CD34+/CD38− CML LSC were found to co-express the homing-receptor CD44, G-CSF-R (CD114), KIT (CD117), FLT3 (CD135), and CXCR4 (CD184). Similar to LSC in AML and CMML, CML LSC were found to display higher levels of Siglec-3 (CD33) and IL-3RA (CD123). Most significantly, however, we found that in contrast to normal CD34+/CD38− stem cells, CD34+/CD38− CML LSC aberrantly express IL-2RA (CD25), dipeptidylpeptidase IV (DPPIV=CD26), and IL-1RAP. In other myeloid leukemias (AML, CMML), CD34+/CD38− LSC also co-expressed CD25, but usually did not express CD26 or IL-1RAP. Whereas CD26 was expressed almost invariably on CD34+/CD38− cells in all CML patients tested, the surface enzyme was neither detectable in more mature CD34+/CD38+ progenitor cells nor on CD34+/CD38− stem cells in reactive bone marrow or healthy controls. During successful treatment with imatinib or nilotinib (patients examined at CCyR and/or MMR), CD34+/CD38− stem cells invariably showed a ‘normal' phenotype (CD25−, CD26−, IL-1RAP−), whereas in relapsing CML, CD34+/CD38− cells were again found to co-express CD25 and CD26. Sorted Lin−/CD26− stem cells obtained from CML patients (at diagnosis) engrafted irradiated NOD-SCID IL-2Rγ−/− (NSG) mice with normal multilineage BCR/ABL1− hematopoiesis, whereas Lin−/CD26+ stem cells were found to engraft NSG mice with BCR/ABL+ cells. We next examined the regulation of expression of CD25 and CD26 on CML LSC. Whereas expression of CD25 was found to depend on BCR/ABL1 and STAT5-activity, CD26 expression was found to be expressed independent of BCR/ABL1 and independent of STAT5-signaling. In a next step, we examined the potential function of CD26 on CML LSC. In these studies, CD26 was identified as a target-enzyme disrupting the niche-related SDF-1α/CXCR4 axis by degrading SDF-1α. Correspondingly, CD26-targeting gliptins (sitagliptin, 1 μM; vildagliptin, 1 μM) were found to revert recombinant DPPIV/CD26-induced or cellular CD26-induced inhibition of SDF-1α-mediated in vitro migration of CD26+ leukemic cells. Finally, we found that in a CML patient treated with nilotinib, in whom uncontrolled diabetes mellitus required therapy with saxagliptin, BCR/ABL1 levels (in percent of ABL according to IS) that were found to increase before the start of saxagliptin (IS before saxagliptin: 1.6 [-4 months], 2.3 [-3 months] , and 2.4 [at therapy-start]), decreased over time during saxagliptin-therapy (IS: 1.0 [+1 month] , 1.0 [+3 months], 0.8 [+5 months] ). Together, the CML-initiating LSC is a CD34+/CD38− cell that exhibits aberrant expression of IL-1RAP, CD25, and DPPIV/CD26. All three markers may be useful for purification of CML LSC. DPPIV/CD26 appears to be a functionally and pathogenetically relevant antigen that may facilitate niche-independent uncontrolled redistribution and thus extramedullary spread of LSC and LSC-derived progenitor cells in CML. Whether CD26 can be developed as a novel therapeutic target in CML is currently under investigation. Disclosures: Valent: Novartis: Consultancy, Honoraria, Research Funding; Bristol-Myers Squibb: Consultancy, Honoraria, 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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3484-3484
    Abstract: Abstract 3484 Acute myeloid leukemia (AML) is a stem cell-derived malignancy characterized by uncontrolled proliferation and accumulation of myeloblasts in hematopoietic tissues. The clinical course and prognosis in AML vary depending on deregulated genes, cell type(s) involved, and the biological properties of the clone. In most variants of AML, the complexity and heterogeneity of oncogenomes pose a challenge for the development of effective targeted therapeutics. However, diverse genetic aberrations in AML typically converge functionally to dysregulate the same cellular core processes. One key event is the corruption of myeloid cell-fate programs resulting in the generation of aberrantly self-renewing leukemia stem cells (LSC), which maintain and propagate the disease and are often resistant to conventional chemotherapy. Hence, strategies aimed at terminating aberrant self-renewal and eradicating LSC are considered as key for the development of more effective AML therapies. In an effort to systematically probe genes involved in chromatin regulation as potential therapeutic targets, we recently employed an unbiased screening approach combining AML mouse models and new in-vivo RNAi technologies, through which we identified the epigenetic ‘reader' BRD4 as new candidate drug target in AML (Zuber et al., Nature, in press). Inhibition of BRD4 using RNAi or a new small-molecule inhibitor (JQ1) blocking BRD4 binding to acetylated histones, showed profound antileukemic effects in AML mouse models, in all human AML cell lines tested (n=8) as well as in primary AML cells. In all models tested, BRD4 suppression was found to trigger apoptosis as well as terminal myeloid differentiation, and potently suppressed expression programs previously associated with LSC. As one key target, we observed a dramatic transcriptional repression of MYC, which recently has been discussed as core component of an LSC associated transcriptional module. To further evaluate suppression of BRD4 as a potential therapeutic approach to eradicate LSC in human AML, we analyzed the effects of JQ1 in primary AML cells obtained from 17 patients with freshly diagnosed or relapsed/refractory AML (females, n=5, males, n=12, median age: 54 years; range: 21–80 years). In unfractionated primary AML cells, submicromolar doses of JQ1 were found to induce major growth-inhibitory effects (IC50 between 0.05 and 0.5 μM) in a broad spectrum of AML subtypes. No differences in IC50 values were seen when comparing drug effects in AML cells kept in the presence or absence of growth-stimulating cytokines (G-CSF, IL-3, SCF). In addition, JQ1 treatment effectively triggered apoptosis in all patients tested, with similar anti-leukemic activities observed in newly diagnosed pts and refractory/relapsed AML. To further evaluate the clinical value of BRD4 as a clinically relevant target in AML, we analyzed the effect of JQ1 on AML LSC. In these experiments, JQ1 effectively induced apoptosis in CD34+/CD38+ progenitor cells as well as in CD34+/CD38− AML stem cells in all donors examined as evidenced by combined surface/Annexin-V staining. Furthermore, JQ1 was found to induce morphologic signs of maturation in 6 of 7 patients examined, thereby confirming our previous data obtained in mouse AML cells. Finally, we were able to show that JQ1 synergizes with Ara-C in inducing growth inhibition in HL60 cells and KG-1 cells. In summary, our data show that small-molecule inhibition of BRD4 has strong anti-leukemic effects in a broad range of AML subtypes. Furthermore, our results support the notion that JQ1's ability to suppress LSC specific transcriptional modules may translate into a therapeutic entry point for eradicating LSC in primary AML. While a more extensive in vivo evaluation of these effects, as well as the development of pharmacologically improved compounds will be required, all existing data unambiguously highlight small-molecule inhibition of BRD4 as a new promising concept in AML therapy. 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: 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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1575-1575
    Abstract: Abstract 1575 The concept of leukemic stem cells (LSC) is increasingly employed to explain the biology of various myeloid neoplasms and to screen for pivotal targets, with the hope to improve drug therapy through elimination of disease-initiating cells. Although the stem cell hypothesis may apply to all neoplasms, leukemia-initiating cells have so far only been characterized in some detail in myeloid leukemias. In an attempt to identify novel cell surface markers and targets on leukemic stem cells (LSC) in acute (AML) and chronic myeloid leukemia (CML), we examined CD34+/CD38- and CD34+/CD38+ populations of leukemic cells in a cohort of patients with AML (n=55) and CML (n=20). In a first step, cell surface antigen profiles were determined by multicolor flow cytometry. In this screen, we were able to show that CD34+/CD38- LSC in AML and CML consistently express certain cytokine receptors, including G-CSFR (CD114), SCFR/KIT (CD117), and IL-3RA (CD123). The low affinity IL-2R (CD25) was detectable on CD34+/CD38- stem cells in patients with CML, and in a subset of AML patients. Other cytokine receptors (R) such as FLT3, IGF-1R, endoglin (CD105), GM-CSFRA (CD116), and OSMR were expressed variably on CD34+/CD38- progenitor cells, whereas the EPOR was not detectable on LSC. We were also able to detect several established therapeutic targets on LSC, including CD33 and CD44. Whereas CD44 was consistently expressed on all LSC in all donors, CD33 was found to be expressed variably on subpopulations of LSC in AML and CML, depending on the phase and type of disease. By using cytokine ligands (G-CSF, IL-3, SCF, EPO) and targeted drugs, we were also able to confirm that identified cytokine receptors and targets were functionally active molecules and potentially relevant targets. In a next step, highly enriched (purity 〉 98%) sorted CD34+/CD38- cells, CD34+/CD38+ cells, and CD34- cells were collected in patients with AML and CML, and in 3 cord blood samples as controls. Purified cells were subjected to gene chip analyses, qPCR, and functional analyses. The identity of leukemic progenitors was confirmed by FISH, and expression of markers and targets in CML stem cells and AML stem cells was confirmed by qPCR. In gene chip analyses, we screened for novel LSC markers and targets. Candidate genes were selected based and their specific expression in progenitor cell fractions and surface membrane location, which was confirmed by antibody staining experiments. Novel stem cell markers identified so far include ROBO4, NPDC-1, and CXCR7. The previously described surface markers MDR-1 and CLL-1 were also identified by flow cytometry, but were also found to be expressed on more mature hematopoietic cells. By contrast, ROBO4 was found to be expressed preferentially on CD34+/CD38- stem cells, but less abundantly on CD34+/CD38+ progenitor cells in CML. Interestingly, whereas ROBO4 was expressed on CD34+/CD38- stem cells in most patients with CML, ROBO4 expression on leukemic stem cells was only found in a subset of AML patients. By contrast, CD34+/CD38- stem cells in AML frequently expressed CLL-1 and NPDC-1 on their surface. In conclusion, we have identified novel markers and targets in CD34+/CD38- progenitor cells in AML and CML. These markers may be useful for the identification and isolation of leukemic stem cells in AML and CML, and for the validation of drug effects on these cells. Disclosures: De Angelis: Biopharm R & D, GSK: Employment. Holmes:Biopharm R & D, GSK: Employment. Valent:Domantis: Research Funding.
    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 ...
  • 10
    In: Oncotarget, Impact Journals, LLC, Vol. 3, No. 12 ( 2012-12-31), p. 1588-1599
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2012
    detail.hit.zdb_id: 2560162-3
    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