Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (10)
  • 1
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 17, No. 5 ( 2018-05-01), p. 977-987
    Abstract: We previously demonstrated that miR-29b-3p is a hopeful miRNA-based therapy against colorectal cancer. In this study, we aimed to clarify a value of miR-29b-1-5p as a next-generation treatment, especially for KRAS-mutant colorectal cancer. RT-PCR assay showed that the expression of miR-29b-3p was high, and its partner strand, miR-29b-1-5p, level was only negligible in clinical colorectal cancer samples. Mimic-miR-29b-1-5p significantly inhibited proliferation of KRAS-mutant colorectal cancer cell lines DLD1 and SW480 and KRAS wild-type HT29 cells. Proliferative activity was further examined by either miR-29b-1-5p strand or its opposite complementary sequence because miR-29b-1-5p is a passenger miRNA and may have no physiologic function. We found that completely opposite complementary strand to miR-29b-1-5p, but not miR-29b-1-5p, possessed a potent antitumor effect and named this byproduct miRNA sequence “MIRTX.” MIRTX directly targeted the 3′-UTR of CXCR2 and PIK3R1 mRNA and suppressed the NF-κB signaling pathway in KRAS-mutated colorectal cancer cells. MIRTX induced apoptosis in DLD1 with downregulation of antiapoptotic BCL2, BCL-xL, and MCL1 and upregulation of cleaved caspase-3 and cleaved PARP. In mouse xenograft models, systemic administration of MIRTX using a super carbonate apatite as a delivery vehicle significantly inhibited tumor growth of DLD1 and HT29 cells without any particular toxicities. In conclusion, these findings indicate that inhibition of NF-κB signaling by this novel miRNA-based therapeutic could be a promising treatment against refractory KRAS-mutant colorectal cancer and KRAS wild-type colorectal cancer. Mol Cancer Ther; 17(5); 977–87. ©2018 AACR.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    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. 79, No. 8 ( 2019-04-15), p. 2031-2041
    Abstract: The oncogenic MUC1-C protein is overexpressed in triple-negative breast cancer (TNBC) cells and contributes to their epigenetic reprogramming and chemoresistance. Here we show that targeting MUC1-C genetically or pharmacologically with the GO-203 inhibitor, which blocks MUC1-C nuclear localization, induced DNA double-strand breaks and potentiated cisplatin (CDDP)-induced DNA damage and death. MUC1-C regulated nuclear localization of the polycomb group proteins BMI1 and EZH2, which formed complexes with PARP1 during the DNA damage response. Targeting MUC1-C downregulated BMI1-induced H2A ubiquitylation, EZH2-driven H3K27 trimethylation, and activation of PARP1. As a result, treatment with GO-203 synergistically sensitized both mutant and wild-type BRCA1 TNBC cells to the PARP inhibitor olaparib. These findings uncover a role for MUC1-C in the regulation of PARP1 and identify a therapeutic strategy for enhancing the effectiveness of PARP inhibitors against TNBC. Significance: These findings demonstrate that targeting MUC1-C disrupts epigenetics of the PARP1 complex, inhibits PARP1 activity, and is synergistic with olaparib in TNBC cells.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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. 79, No. 13_Supplement ( 2019-07-01), p. 3707-3707
    Abstract: Primary cultured cells derived from an individual patient’s tumor have phenotypic heterogeneity unlike established cancer cell lines. We established organoid in two-dimensional (2D) culture with the reproducibility of clinical phenotypic heterogeneity as a human tumor model, named isolated tumor-derived cancer cells (iCCs). The success rate of iCC growth in vitro was 100%, passage was 90%, and establishment of a xenograft model from iCC was 80%. Established 2DO can proliferate well, and are easier to use in several assays compared with reported organoid in 3D culture. However, established 2DO also can be easily cultured in a 3D culture system. iCC populations were analyzed by flow cytometry. Most iCCs expressed epithelial cell adhesion molecule (EpCAM), and the high- and low-expressing CD44 groups were identified. Seral markers were examined, and we reviled that TRA1-81-positive cells in the high-expressing CD44 group were necessary for replicating clinical tumor heterogeneity. Moreover, basic fibroblast growth factor (bFGF) was necessary to keep TRA1-81-positive cells. Next, we did anticancer drug assay using iCCs from 10 patients and predicted the clinical efficacy of drugs. Six patients received chemotherapy after their surgery, and four patients of them had distant metastases. The in vitro sensitivity was compared to clinical outcomes (RECIST criteria) in four patients that all had distant metastases. The concentration of anti-cancer drugs was set as estimated concentration in human tissue. In the examination using 5-FU and oxaliplatin, the survival rates of iCCs were more than 85% in one progressive disease (PD) patient. The survival rates of iCCs were less than 83% in three stable disease (SD) patients (median, 70%). The survival rates were also less than 83% in two patients who underwent adjuvant chemotherapy with no recurrence (median 70%). After 3 months chemotherapy, PD patient’s cell survival rates were more than 85%, and SD patient’s cell survival rates were less than 83%. After 6 months chemotherapy, PD patient’s cell survival rates were more than 81%, and SD patient’s cell survival rates were less than 75%. Therefore, the cut-off value was considered to be 83-85% for 3 months to keep SD, and 75-80% for 6 months. However, more examination is need to develop a prediction model we can use as a clinical application. In conclusion, our primary culture model may be a novel tool as a human tumor model with tumor heterogeneity, and it will be used to predict therapeutic effect of anti-cancer drugs in the clinical field. Citation Format: Shiki Fujino, Norikatsu Miyoshi, Kazuhiro Saso, Masaru Sasaki, Masayoshi Yasui, Masayuki Ohue, HIdekazu Takahashi, Naotsugu Haraguchi, Chu Matsuda, Taishi Hata, Tsunekazu Mizushima, Masaki Mori, Yuichiro Doki. 2D organoid (2DO) as human tumor model for predicting therapeutic effect of anti-cancer drugs [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3707.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 66, No. 2 ( 2006-01-15), p. 812-819
    Abstract: c-Maf translocation or overexpression has been observed in human multiple myeloma. Although c-maf might function as an oncogene in multiple myeloma, a role for this gene in other cancers has not been shown. In this study, we have found that mice transgenic for c-Maf whose expression was direct to the T-cell compartment developed T-cell lymphoma. Moreover, we showed that cyclin D2, integrin β7, and ARK5 were up-regulated in c-Maf transgenic lymphoma cells. Furthermore, 60% of human T-cell lymphomas (11 of 18 cases), classified as angioimmunoblastic T-cell lymphoma, were found to express c-Maf. These results suggest that c-Maf might cause a type of T-cell lymphoma in both mice and humans and that ARK5, in addition to cyclin D2 and integrin β7, might be downstream target genes of c-Maf leading to malignant transformation. (Cancer Res 2006; 66(2): 812-9)
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2006
    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 ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 4397-4397
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 4397-4397
    Abstract: Background: The anti-epidermal growth factor receptor (EGFR) monoclonal antibodies are the molecular target drugs, dependent on v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations status in metastatic colorectal cancer (mCRC). Anti-EGFR monoclonal antibody based chemotherapy is the standard treatment for mCRC with KRAS wild-type. However, the issue of resistance to this therapy in CRC with KRAS mutations (approximately 50% of CRC) remains unresolved. The aim of this study is to identify microRNAs (miRNAs) that govern the resistance to anti-EGFR monoclonal antibody in CRC with KRAS mutations. Experimental Design: We first established the KRAS mutant cell model using HEK293 and MRC5 cells by introducing KRASG12V plasmid. The miRNA expression profiles were compared between these cells using miRNA microarray analysis. We selected the candidate a miRNA which down-regulates the downstream signal of KRAS using luciferase reporter assays. Assessment of target genes of the candidate a miRNA was performed in vitro. Next, anti-tumor effects by supplement of a miRNA were evaluated in vitro and in vivo. Results: We picked up miRNAs, of which expressions decreased in both HEK293 and MRC5 cells by introduction of KRASG12V plasmid in miRNA array analysis. Among them, administration of mature-miR-X suppressed SRE and AP1 activity in KRAS mutant CRC cell lines such as DLD1G13D, SW480 G12V, and HCT116 G13D using dual luciferase reporter assay. Furthermore, miR-X regulated not only Ras/Raf/MAPK pathway but also suppressed PI3K/Akt pathway in vitro. Administration of mature-miR-X suppressed proliferation and induced apoptosis of the CRC cell lines with KRAS mutations in vitro. Systemic mature-miR-X administration into tail veins of nude mice significantly inhibited the growth of pre-established DLD1G13D xenografts. miR-X level was significantly lower in tumor tissues compared to normal mucosa. Conclusions: miR-X may overcome anti-EGFR therapy resistance in CRC with KRAS mutations by regulating Ras/Raf/MAPK pathway and PI3K/Akt pathway. Our data suggests that miR-X could be suitable for a novel therapy for mCRC patients with KRAS mutations. Citation Format: Masayuki Hiraki, Junichi Nishimura, Mamoru Uemura, Taishi Hata, Ichiro Takemasa, Tsunekazu Mizushima, Hirofumi Yamamoto, Yuichiro Doki, Masaki Mori. Therapeutic microRNA agaisnt KRAS mutant colorectal cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4397. doi:10.1158/1538-7445.AM2014-4397
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Molecular Cancer Therapeutics Vol. 18, No. 10 ( 2019-10-01), p. 1744-1754
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 10 ( 2019-10-01), p. 1744-1754
    Abstract: The oncogenic MUC1-C protein and the TWIST1 epithelial–mesenchymal transition transcription factor (EMT-TF) are aberrantly expressed in triple-negative breast cancer (TNBC) cells. However, there is no known association between MUC1-C and TWIST1 in TNBC or other cancer cells. Here, we show that MUC1-C activates STAT3, and that MUC1-C and pSTAT3 drive induction of the TWIST1 gene. In turn, MUC1-C binds directly to TWIST1, and MUC1-C/TWIST1 complexes activate MUC1-C expression in an autoinductive circuit. The functional significance of the MUC1-C/TWIST1 circuit is supported by the demonstration that this pathway is sufficient for driving (i) the EMT-TFs, ZEB1 and SNAIL, (ii) multiple genes in the EMT program as determined by RNA-seq, and (iii) the capacity for cell invasion. We also demonstrate that the MUC1-C/TWIST1 circuit drives (i) expression of the stem cell markers SOX2, BMI1, ALDH1, and CD44, (ii) self-renewal capacity, and (iii) tumorigenicity. In concert with these results, we show that MUC1-C and TWIST1 also drive EMT and stemness in association with acquired paclitaxel (PTX) resistance. Of potential therapeutic importance, targeting MUC1-C and thereby TWIST1 reverses the PTX refractory phenotype as evidenced by synergistic activity with PTX against drug-resistant cells. These findings uncover a master role for MUC1-C in driving the induction of TWIST1, EMT, stemness, and drug resistance, and support MUC1-C as a highly attractive target for inhibiting TNBC plasticity and progression.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 3426-3426
    Abstract: Incidence rates of Wilms tumor (WT) markedly differ among ethnic groups; the epidemiological studies showed that the incidence of WT in Japanese children is about half of that of WT in Caucasian children. In Hawaii and Britain, the rates of WT in Asian descent were less than two-third or a half of those in Caucasian children, and these findings suggest that the different incidences of WT may be caused by genetic factor(s), but not by environmental factor(s). Development of WT is associated with mutations of certain genes, including WT1, WTX and CTNNB1, and abnormal expression of IGF2, namely, loss of imprinting (LOI). The frequency of these abnormalities occurs in 10-15% (WT1), 7-15% (WTX), 5-13% (CTNNB1) and 30-60% (IGF2 LOI) of sporadic WTs based on the studies in North-American or European children. To demonstrate the molecular characteristic in Japanese WT, we examined mutations/deletions of WT1, WTX and CTNNB1 using Southern blot, SNP array, sequencing and/or real-time PCR, and expression pattern of IGF2 using the ApaI polymorphism site in exon 9 and the methylation status of the CTCF binding site 6 at H19-differential methylated region (DMR) in 101 sporadic and 13 syndromic WTs in Japanese children. Abnormalities of WT1, WTX or CTNNB1 were detected in 24 (23.8%), 24 (23.8%), and 21 (20.8%)of 101 sporadic WTs, respectively, and in 13 (100%), 0, and 10 (76.9%) of 13 syndromic WTs, respectively. LOI of IGF2 was found in 24 (23.8%) of sporadic WTs and in 1 (7.7%) of syndromic WTs. Thus, the incidence of WT1, WTX or CTNNB1 abnormality was higher, but that of LOI of IGF2 was lower in Japanese children than in Caucasian counterparts. Because the incidence of WT in Japanese children is about half of that of WT in Caucasian ones, the population-based incidences of WT1, WTX or CTNNB1 abnormality may be similar between the two ethnic groups. In contrast, the incidence rate of IGF2-LOI in Japanese children was lower than those of IGF2-LOI in Caucasian children, and the difference in the population-based incidences is much larger. A recent study of 47 Japanese patients with Beckwith-Wiedemann syndrome revealed a significantly lower frequency of H19-DMR hypermethylation in Japanese patients than in North American and European patients (Sasaki et al, Eur J Hum Genet 2007). This and the present findings suggest that susceptibility to epigenetic alterations differs between Japanese and Caucasian populations. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3426.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    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 ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 2930-2930
    Abstract: Primary culture of cancer cells derived from each patient’s tumor can provide important information of the “individual tumor.” It is general to use cell lines in basic research filed. However, cell lines are quite different from clinical cancers. Clinical cancer tissues are composed of not only cancer cells but also tumor microenvironments such as stromal cells and tumor vessels. The primary culture method of clinical cancer with tumor niche microenvironments has not been optimized. We have developed a simple 2D-culture method for primary colorectal cancer (CRC). We obtained 30 samples from surgically resected tumor. They were mechanically and enzymatically digested and fibrotic tissue and bacteria were excluded using customized two size filters. And we cultured the obtained cells on a matrigel-coated plate with embryonic stem (ES) cells culture medium. We named these cultured cancer cells, “isolated-tumor derived Cancer Cells (iCCs).” All iCCs grew and about 80 % of iCCs were successfully passaged. Twenty-three iCCs were transplanted into the subcutaneous layer of NOD-SCID mice, and the tumor growth and histology of iCCs were examined. The morphology was similar to each parental clinical tumor. And microarray analysis showed that RNA expression of iCCs was similar to each parental tumor. Furthermore, we examined the culture medium; our modified ES culture medium (ES-cultured iCCs) and 10% FBS medium (serum-cultured iCCs). The expression of surface markers regarding cancer stem cells such as CD44 and CD24 were different between ES-cultured iCCs and serum-cultured iCCs, and drug sensitivity of iCCs were also different. FACS analysis and immunocytochemistry revealed that iCCs contained PDGFR-positive cells. The results of multi-drug sensitivity assay were different between iCCs and cell lines especially in PDGFR inhibitor. Strong correlations were observed between the results of multi-drug sensitivity assay of iCCs and clinical outcomes of chemotherapy. We report an innovative primary culture method and the in vivo and in vitro analyses of iCCs, leading to the future application. Citation Format: Shiki Fujino, Norikatsu Miyoshi, Masayuki Ohue, Yusuke Takayashi, Masayoshi Yasui, Hidekazu Takahashi, Naoyuki Haraguchi, Jhunichi Nishimura, Taishi Hata, Tsunekazu Mizushima, Yuichiro Doki, Masaki Mori. Niche microenvironments essential for tumor heterogeneity of primary cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2930. doi:10.1158/1538-7445.AM2017-2930
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 1 ( 2018-01-01), p. 205-215
    Abstract: The immune checkpoint ligand PD-L1 and the transmembrane mucin MUC1 are upregulated in triple-negative breast cancer (TNBC), where they contribute to its aggressive pathogenesis. Here, we report that genetic or pharmacological targeting of the oncogenic MUC1 subunit MUC1-C is sufficient to suppress PD-L1 expression in TNBC cells. Mechanistic investigations showed that MUC1-C acted to elevate PD-L1 transcription by recruitment of MYC and NF-κB p65 to the PD-L1 promoter. In an immunocompetent model of TNBC in which Eo771/MUC1-C cells were engrafted into MUC1 transgenic mice, we showed that targeting MUC1-C associated with PD-L1 suppression, increases in tumor-infiltrating CD8+ T cells and tumor cell killing. MUC1 expression in TNBCs also correlated inversely with CD8, CD69, and GZMB, and downregulation of these markers associated with decreased survival. Taken together, our findings show how MUC1 contributes to immune escape in TNBC, and they offer a rationale to target MUC1-C as a novel immunotherapeutic approach for TNBC treatment. Significance: These findings show how upregulation of the transmembrane mucin MUC1 contributes to immune escape in an aggressive form of breast cancer, with potential implications for a novel immunotherapeutic approach. Cancer Res; 78(1); 205–15. ©2017 AACR.
    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 ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 22 ( 2019-11-15), p. 5711-5722
    Abstract: The NuRD chromatin remodeling and deacetylation complex, which includes MTA1, MBD3, CHD4, and HDAC1 among other components, is of importance for development and cancer progression. The oncogenic mucin 1 (MUC1) C-terminal subunit (MUC1-C) protein activates EZH2 and BMI1 in the epigenetic reprogramming of triple-negative breast cancer (TNBC). However, there is no known link between MUC1-C and chromatin remodeling complexes. Here, we showed that MUC1-C binds directly to the MYC HLH-LZ domain and identified a previously unrecognized MUC1-C→MYC pathway that regulates the NuRD complex. MUC1-C/MYC complexes selectively activated the MTA1 and MBD3 genes and posttranscriptionally induced CHD4 expression in basal- but not luminal-type BC cells. In turn, MUC1-C formed complexes with these NuRD components on the ESR1 promoter. Downregulating MUC1-C decreased MTA1/MBD3/CHD4/HDAC1 occupancy and increased H3K27 acetylation on the ESR1 promoter, with induction of ESR1 expression and downstream estrogen response pathways. Targeting MUC1-C and these NuRD components also induced expression of FOXA1, GATA3, and other markers associated with the luminal phenotype. These findings support a model in which MUC1-C activates the NuRD complex to drive dedifferentiation and reprogramming of TNBC cells. Significance: MUC1-C directly interacts with MYC to activate the NuRD complex, mediating regulation of the estrogen receptor in triple-negative breast cancer cells.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages