Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
  • Li, Yan  (26,209)
Type of Medium
Person/Organisation
Language
Years
  • 1
    In: BMC Cancer, Springer Science and Business Media LLC, Vol. 18, No. 1 ( 2018-12)
    Type of Medium: Online Resource
    ISSN: 1471-2407
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2041352-X
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 4491-4491
    Abstract: Abstract 4491 Background: Mantle cell lymphoma is typically considered to be aggressive and incurable. About 15% of these patients have an indolent course. MCL demonstrates the aggressive features of a rapidly progressive neoplasm but with the negative consequences of an indolent lymphoma, namely incurability and frequent relapses. The median overall survival (OS) was reported at 3–4 years when MCL was first described in the 1990's. OS has since increased substantially and this is thought to be secondary to more aggressive initial therapy and improvement in supportive care. In many parts of the world, autologous stem cell transplant (ASCT) is incorporated in the front line therapy for MCL patients with good performance status. However, improvement in survival has not deemed MCL a curable disease. Concern for treatment-related morbidity seen with aggressive therapy in an incurable disease has led some centers to practice a more conservative approach. The purpose of our study was to compare patient characteristics and the overall survival of patients treated aggressively with ASCT versus conservatively with either conventional chemotherapy or no treatment at a single institution. Methods: 52 cases of confirmed mantle cell lymphoma diagnosed at Rush University Medical Center between January 2000 and November 2010 were studied. Demographic, clinical and treatment data were collected and reviewed. The Social Security Death Index and hospital records were used to assess survival. Comparative survival analysis was performed based on treatment strategies including the following: no treatment (watch and wait), chemotherapy, ASCT at any time during course of treatment. None of these patients had an allogeneic stem cell transplant. Results: 43 of the 52 cases met all inclusion criteria and had complete diagnostic and treatment data. The no-treatment group consisted of 5 cases with a median age of 59 years. The chemotherapy group included 23 cases with a median age of 68 years. The most common initial therapy was RCHOP in 14 cases, followed by various other regimens (i.e. bortezomib + rituximab, bendamustine + rituximab) in 7 cases and HyperCVAD in 2 cases. The ASCT group included 15 cases with a median age of 61 years. Pre-transplant chemotherapy was RCHOP in 4 cases, HyperCVAD in 5 cases and other regimens in 6 cases. The comparative survival analysis for the three treatment groups was not statistically significant (p=0.496) and the estimated 3 year OS was 100% for the no treatment group, 74% for the chemotherapy group and 85% for the ASCT group. The estimated 5 year OS was 100% for the no treatment group, 66% for the chemotherapy group and 68% for the ASCT group. There were no cases of allogeneic stem cell transplants. Conclusions: Our review of MCL cases treated at a single institution supports a role for conservative treatment approaches to this disease entity. This can avoid the potential long term morbidity from ASCT in a subgroup of patients while still keeping the modality of therapy as an option for them at relapse. The incidence of indolent MCL requiring no treatment was 12% which is consistent with those seen in other studies. Further research is necessary to guide treatment decisions for MCL patients whose disease characteristics are intermediate between aggressive and indolent. Disclosures: Gregory: Genentech:.
    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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 4178-4178
    Abstract: Abstract 4178 Fanconi anemia (FA) is a rare recessive disease manifested by progressive bone marrow (BM) failure, congenital abnormalities and a predisposition to cancer. The only curative treatment for the pancytopenia to date is allogeneic stem cell transplantation. However, because allogeneic grafts are available to only about 30% of patients and mismatch grafts have a higher incidence of sequelae, autologous gene transfer into hematopoietic stem cells (HSCs) is a potential viable therapy. Major obstacles for gene therapy in FA include: low numbers of HSCs, an increased frequency of apoptosis and propensity for clonal selection following prolonged culture and the risks of all gene transfer technology including transactivation of proto-oncogenes from potent viral promoters such as the spleen focus-forming virus promoter (SFFV). We have recently found that a lentiviral (LV) vector containing SFFV promotor rendered a high functional and biochemical expression of the transgene in human and murine repopulating HSCs. However, given the recent concerns in the use of potent viral promoters like SFFV, in this study we developed LV vectors harboring a mammalian cell endogenous promoter human phosphoglycerate kinase (hPGK) to drive the expression of human FANCA and tested the ability of this construct to correct the phenotypes of human FANCA−/− fibroblasts and murine Fanca−/− hematopoietic stem and progenitor cells. Viral-mediated functional correction of FANCA was first evidenced by the restoration of FANCD2 mono-ubiquitination and correction of mitomycin C (MMC)-induced G2/M cell cycle arrest in human FANCA−/− fibroblast cells. In addition, the survival of genetically modified murine Fanca−/− clonogenic progenitors in the presence of 5nM MMC was improved from 30% (transduced with EGFP reporter) to 80% (transduced with FANCA cDNA), nearly equivalent to the survival of WT progenitors (90%) at that concentration. In addition, in a competitive repopulation assay, the long-term repopulating ability of Fanca−/− stem cells in the lethally-irradiated recipient mice was dramatically increased from 10% test cell chimerism (transduced with EGFP reporter) to 55% test cell chimerism (transduced with FANCA cDNA). Furthermore, the chimerism was maintained over 6 months in the primary recipients and an additional 6 months in the secondary recipients. Finally, resistance to MMC was retained in progenitors cultured from FACS sorted bone marrow test cells from the primary and secondary recipients. Collectively, these data suggest that the lentiviral construct using a mammalian cell endogenous promoter (hPGK) is sufficient to correct the phenotypes of murine FA HSC/progenitor cells. Studies are now ongoing to examine LV mediated FANCA expression in human primitive progenitor/ SCID/NOD repopulating cells. 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 ...
  • 4
    Online Resource
    Online Resource
    Informa UK Limited ; 2011
    In:  Journal of the American Statistical Association Vol. 106, No. 495 ( 2011-09), p. 1067-1074
    In: Journal of the American Statistical Association, Informa UK Limited, Vol. 106, No. 495 ( 2011-09), p. 1067-1074
    Type of Medium: Online Resource
    ISSN: 0162-1459 , 1537-274X
    RVK:
    RVK:
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2011
    detail.hit.zdb_id: 2064981-2
    detail.hit.zdb_id: 207602-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 116, No. 26 ( 2010-12-23), p. 5972-5982
    Abstract: Because primary myelofibrosis (PMF) originates at the level of the pluripotent hematopoietic stem cell (HSC), we examined the effects of various therapeutic agents on the in vitro and in vivo behavior of PMF CD34+ cells. Treatment of PMF CD34+ cells with chromatin-modifying agents (CMAs) but not hydroxyurea, Janus kinase 2 (JAK2) inhibitors, or low doses of interferon-α led to the generation of greater numbers of CD34+ chemokine (C-X-C motif) receptor (CXCR)4+ cells, which were capable of migrating in response to chemokine (C-X-C motif) ligand (CXCL)12 and resulted in a reduction in the proportion of hematopoietic progenitor cells (HPCs) that were JAK2V617F+. Furthermore, sequential treatment of PMF CD34+ cells but not normal CD34+ cells with decitabine (5-aza-2′-deoxycytidine [5azaD]), followed by suberoylanilide hydroxamic acid (SAHA; 5azaD/SAHA), or trichostatin A (5azaD/TSA) resulted in a higher degree of apoptosis. Two to 6 months after the transplantation of CMAs treated JAK2V617F+ PMF CD34+ cells into nonobese diabetic/severe combined immunodeficient (SCID)/IL-2Rγnull mice, the percentage of JAK2V617F/JAK2total in human CD45+ marrow cells was dramatically reduced. These findings suggest that both PMF HPCs, short-term and long-term SCID repopulating cells (SRCs), are JAK2V617F+ and that JAK2V617F+ HPCs and SRCs can be eliminated by sequential treatment with CMAs. Sequential treatment with CMAs, therefore, represents a possible effective means of treating PMF at the level of the malignant SRC.
    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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 113, No. 10 ( 2009-03-05), p. 2342-2351
    Abstract: Fanconi anemia (FA) is a heterogeneous genetic disorder characterized by bone marrow failure and complex congenital anomalies. Although mutations in FA genes result in a characteristic phenotype in the hematopoietic stem/progenitor cells (HSPCs), little is known about the consequences of a nonfunctional FA pathway in other stem/progenitor cell compartments. Given the intense functional interactions between HSPCs and the mesenchymalmicroenvironment, we investigated the FA pathway on the cellular functions of murine mesenchymal stem/progenitor cells (MSPCs) and their interactions with HSPCs in vitro and in vivo. Here, we show that loss of the murine homologue of FANCG (Fancg) results in a defect in MSPC proliferation and in their ability to support the adhesion and engraftment of murine syngeneic HSPCs in vitro or in vivo. Transplantation of wild-type (WT) but not Fancg−/− MSPCs into the tibiae of Fancg−/− recipient mice enhances the HSPC engraftment kinetics, the BM cellularity, and the number of progenitors per tibia of WT HSPCs injected into lethally irradiated Fancg−/− recipients. Collectively, these data show that FA proteins are required in the BM microenvironment to maintain normal hematopoiesis and provide genetic and quantitative evidence that adoptive transfer of WT MSPCs enhances hematopoietic stem cell engraftment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    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
    Online Resource
    Online Resource
    American Society of Hematology ; 2016
    In:  Blood Vol. 128, No. 22 ( 2016-12-02), p. 5559-5559
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 5559-5559
    Abstract: Myelodysplastic syndromes (MDS) are a heterogeneous group of hematological malignancies characterized by a deregulation of blood cell formation with cytopenia in varying degrees and frequently develop into acute myeloid leukemia (AML). Next generation sequencing (NGS) has significantly contributed to diagnosis and prognostication in patients with MDS. To explore the role of acquired mutations in MDS biology and clinical features, we performed mutational profiling of 111 genes in 125 patients with MDS based on target-sequencing. The results showed that 89% of patients had one or more carcinogenic mutations. Mutation frequencies of several genes, including ASXL1 (16.8%), RUNX1 (14.4%) and TET2 (12%), were above 10% in MDS patients. According to univariate analysis in 108 patients with survival data, mutations of GATA1/2, TP53 and DNMT3A were identified as adverse prognostic factors for overall survival (OS), while RUNX1, KRAS/NRAS, SRSF2 and TET2 mutations were determined as unfavorable factors for progression free survival (PFS). By multivariate COX regression analysis, mutations of KRAS/NRAS, GATA1/2 and DNMT3A were independent risk factors for OS, whereas IDH1/2 gene mutations were favorable factors for PFS. By evaluation of the clinical benefit of hypomethylating agents (HMAs), it showed that for patients harboring mutations associated DNA methylation or not and receiving HMAs treatment or not, the order of the outcome from good to bad was geneWT with non-HMAs, geneWT with HMAs, genemutwith HMAs and genemutwith non-HMAs. Therefore, target-sequencing of mutational spectrum is a feasible and highly promising prediction method for prognostic evaluation in patients with MDS, which would contribute to personalized therapeutic decisions. 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: 2016
    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
    Online Resource
    Online Resource
    American Society of Hematology ; 2013
    In:  Blood Vol. 122, No. 21 ( 2013-11-15), p. 5427-5427
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 5427-5427
    Abstract: Bone marrow mesenchymal stem cells (BMSCs) were confirmed that have great potential in decreasing acute graft-versus-host disease(aGVHD) after allogeneic hematopoietic stem cells transplantation(allo-HSCT) in various clinical trails. However, the immuno-modulatory effects of BMSCs are not always successfully achieved in vivo. In this study, we aimed to proved that activation of heme oxygenase-1(HO-1) in BMSCs could significantly enhance the capacity on decreasing aGVHD in vivo and explored the relative mechanism. Methods We cloned mice’s HO-1 cDNA from mice bone marrow and constructed recombinant lenti-virus vectors (Lentivirus-V5-D-TOPO-HO-1-EGFP/Lentivirus-V5-D-TOPO-EGFP), which titer was 1×1011pfu/mL. These mouse BMSCs were separated, cultured, purified, and detected by morphology, flow cytometry, osteogenic, adipogenic and chondrogenic induction. Then recombinant lenti-virus vectors were transferred into mouse BMSCs, and the expression of EGFP and HO-1 were detected by fluorescence microscope, RT-PCR and Western blot respectively. We established mice’s aGVHD model after allo-HSCT. Four groups were separated in vivo test (Group A: aGVHD control; Group B: aGVHD model injected into mouse BMSCs; Group C: aGVHD model injected into mouse BMSCs transfected with EGFP; Group D: aGVHD model injected into mBMSCs transfected with HO-1 gene). The survival time, body weight and clinical score of aGVHD in mice model were monitored. Liver, intestine and lung in each group were obtained for histological examination. Plasma concentrations of interleukin (IL)-2, IL-4, IL-6, IL-10, IFN-γ and TNF-„p were also determined using a Cytometric Bead Array. Results In vitro, over-expression of HO-1 promoted the proliferation of BMSCs, the cell proliferation rate of BMSC transfected with lenti-virus-HO-1 was 1.25 folder higher over natural BMSCs(p 〈 0.05). In vivo, the immuno-suppressive capacity of BMSCs expressing HO-1 in a major histo-compatibility complex (MHC)-mismatched mouse model of bone marrow (BM) transplantation from C57BL/6 donors to BALB/c recipients. Treatment with HO-1 over-expressing BMSCs obviously decreased the animal mortality rate,60% mice in group BMSCs suffered from aGVHD and died, while 70% mice in group BMSCs-HO-1 were still alive, and expressed less clinical and pathological aGVHD scores. In addition, compared with other groups, the level of negative regulative cytokines, IL-2, IL-6, IFN-γ and TNF-„p in recipients injected with BMSCs-HO-1, were significantly decreased (FCBMSC-HO-1/BMSC=cytokines concentration detected in group infused BMSCs-HO-1/group infused BMSCs, if FCBMSC-HO-1/BMSC 〈 1, means the cytokines concentration detected in group infused BMSCs-HO-1 decreased. Our study indicated that: FCBMSC-HO-1/BMSC of IL-2: 0.4±0.1; IL-6: 0.7±0.1; IFN-γ: 0.7±0.05; TNF-„p: 0.8±0.02. p 〈 0.05), while those positively regulative cytokines, IL-4 and IL-10 were increased(FCBMSC-HO-1/BMSC 〉 1, means the cytokines concentration detected in group infused BMSCs-HO-1 increased. Our study indicated that: FCBMSC-HO-1/BMSC of IL-4: 1.3±0.15; IL-10: 1.5±0.1; p 〈 0.05). In field of Th1/Th2 cells proliferation, the value of CD8+ and CD4+ T cells and the ratio of Th1/Th2 T cell subsets decreased(CTBMSC-HO-1/BMSC= cells counts of group infused BMSCs-HO-1/group infused BMSCs, CTBMSC-HO-1/BMSC 〈 1, means cells counts of group infused BMSCs-HO-1 decreased. Our data showed that, CTBMSC-HO-1/BMSC of CD8+ T cells: 0.25±0.09; CD4+ T cells:0.47±0.06; Th1/Th2 T cell subsets: 0.32±0.05; p 〈 0.05) , at the same time the proportion of CD4+CD25+ T cells increased both in spleen lymphocytes in vivo after allo-HSCT with BMSCs expressing HO-1 compared with conventional allo-HSCT(CTBMSC-HO-1/BMSC of CD4+ CD25+ Foxp3+ cells:13.4±2.1; p 〈 0.01. CTBMSC-HO-1/BMSC 〉 1,means cells counts of group infused BMSCs-HO-1 increased). Conclusion our report strongly reveals that activation of HO-1 could enhance the ability of BMSCs to effectively alleviate aGVHD. The mechanism involved that the enhanced homing ability of BMSCs, immuno-suppressive ability to decrease negatively regulative cytokines, while to increase positive part. At last, BMSCs transfected lenti-virus-HO-1 expressed the potential ability to induce Treg cells proliferation. 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: 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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 2993-2993
    Abstract: Abstract 2993 Introduction: Competition from the host hematopoietic compartment is the primary barrier to engraftment after in utero hematopoietic cell transplantation (IUHCT). Prostaglandin E2 (PGE2) has been shown to increase donor cell homing, cell survival, and engraftment after postnatal HCT through increased CXCR4 and Survivin expression. We have previously shown that pre-incubation with Diprotin A, a CD26 inhibitor, confers a homing and engraftment advantage for donor cells after IUHCT. We hypothesize that pre-incubation of donor cells with PGE2 alone, or in combination with Diprotin A, will provide a selective donor cell homing and competitive advantage and enhance long-term engraftment after IUHCT. Methods: Balb/c pregnant mice underwent IUHCT with 1 × 107 B6GFP bone marrow (BM) cells, with or without pre-incubation with PGE2, Diprotin A, or PGE2 plus Diprotin A, by vitelline vein injection at E14. A 1μM per 1×106 BM cell concentration of dmPGE2, a long acting derivative, was used to treat the donor BM for two hours prior to IUHCT. In the combined therapy group, a 50 μM concentration of Diprotin A was used to treat the BM for an additional 15 minutes following PGE2 incubation. Homing to and engraftment in the fetal liver (FL) at early prenatal time points and frequency of donor cells in peripheral blood (PB) at 1 month and PB and BM at 4 months were assessed by flow cytometry. Analysis of donor cell cycling and cell survival was performed by in vitro BrdU cell labeling and anti-survivin and anti-caspase 3 antibody staining respectively. Results: Ex-vivo incubation of donor cells with PGE2 alone, Diprotin A, and PGE2 plus Diprotin A increased homing of allogeneic BM to the FL at 4 and 24 hours after IUHCT with the combined treatment group reaching statistical significance. Analysis of cell cycling of donor cells engrafted in the fetal liver at 96 hours after IUHCT demonstrated a 1.4 fold increase in the proportion of cells in the S and G2+M phases in PGE2 treated cells relative to non-treated BM controls (31.4% vs 21.2%) and a 1.2 fold increase over E18 FL controls (25.5%). A concurrent four fold increase in anti-survivin staining and a seven fold decrease in anti-caspase 3 staining were also seen in PGE2 treated cells engrafted in the fetal liver relative to non-PGE2 treated cells (anti-survivin: 19.8% vs. 4.75%. anti-caspase 3: 9.3% vs. 64.1%). Most importantly, a sustained increase in multi-lineage donor cell chimerism was observed (25–32%) in the PB and BM of recipients of PGE2 treated cells, as compared to controls, which demonstrated a decline from 13.4% to 4.9% over a 4 month period after IUHCT. Treatment with Diprotin A alone demonstrated a smaller increase in donor cell chimerism at 4 months after IUHCT. However, there was no statistical difference at 4 months between the PGE2 and PGE2 plus Diprotin A treatment groups (Figure 1). Conclusions: PGE2 exposure results in markedly improved long-term engraftment of donor cells. This improvement is associated with increased cell cycling, increased survivin expression and reduced apoptosis suggesting that PGE2 exposure confers a competitive advantage to donor hematopoietic stem cells that persists over time by increasing cell cycling and survival. These increases likely explain the long-term advantage of PGE2 treatment. While CD26 blockade conferred an improvement in early homing kinetics, the combination of PGE2 plus CD26 blockade did not result in higher long term engraftment than PGE2 alone. We conclude that PGE2 pre-incubation is a promising method to achieve sustained increased donor cell engraftment after IUHCT. * p 〈 0.05 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 ...
  • 10
    In: BMC Genetics, Springer Science and Business Media LLC, Vol. 21, No. 1 ( 2020-12)
    Abstract: RNA-sequencing was performed to explore the bovine liver transcriptomes of Holstein cows to detect potential functional genes related to lactation and milk composition traits in dairy cattle. The bovine transcriptomes of the nine liver samples from three Holstein cows during dry period (50-d prepartum), early lactation (10-d postpartum), and peak of lactation (60-d postpartum) were sequenced using the Illumina HiSeq 2500 platform. Results A total of 204, 147 and 81 differentially expressed genes (DEGs, p   〈  0.05, false discovery rate q  〈  0.05) were detected in early lactation vs. dry period, peak of lactation vs. dry period, and peak of lactation vs. early lactation comparison groups, respectively. Gene ontology and KEGG pathway analysis showed that these DEGs were significantly enriched in specific biological processes related to metabolic and biosynthetic and signaling pathways of PPAR, AMPK and p53 ( p   〈  0.05). Ten genes were identified as promising candidates affecting milk yield, milk protein and fat traits in dairy cattle by using an integrated analysis of differential gene expression, previously reported quantitative trait loci (QTL), data from genome-wide association studies (GWAS), and biological function information. These genes were APOC2 , PPP1R3B , PKLR , ODC1 , DUSP1 , LMNA, GALE , ANGPTL4 , LPIN1 and CDKN1A . Conclusion This study explored the complexity of the liver transcriptome across three lactation periods in dairy cattle by performing RNA sequencing. Integrated analysis of DEGs and reported QTL and GWAS data allowed us to find ten key candidate genes influencing milk production traits.
    Type of Medium: Online Resource
    ISSN: 1471-2156
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2041497-3
    detail.hit.zdb_id: 3058779-7
    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