GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Cancer Cell, Elsevier BV, Vol. 32, No. 4 ( 2017-10), p. 460-473.e6
    Type of Medium: Online Resource
    ISSN: 1535-6108
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    detail.hit.zdb_id: 2074034-7
    detail.hit.zdb_id: 2078448-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Discovery Vol. 11, No. 3 ( 2021-03-01), p. 660-677
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 11, No. 3 ( 2021-03-01), p. 660-677
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease with a 5-year survival rate of approximately 9%. An improved understanding of PDAC initiation and progression is paramount for discovering strategies to better detect and combat this disease. Although transcriptomic analyses have uncovered distinct molecular subtypes of human PDAC, the factors that influence subtype development remain unclear. Here, we interrogate the impact of cell of origin and different Trp53 alleles on tumor evolution, using a panel of tractable genetically engineered mouse models. Oncogenic KRAS expression, coupled with Trp53 deletion or point mutation, drives PDAC from both acinar and ductal cells. Gene-expression analysis reveals further that ductal cell–derived and acinar cell–derived tumor signatures are enriched in basal-like and classical subtypes of human PDAC, respectively. These findings highlight cell of origin as one factor that influences PDAC molecular subtypes and provide insight into the fundamental impact that the very earliest events in carcinogenesis can have on cancer evolution. Significance: Although human PDAC has been classified into different molecular subtypes, the etiology of these distinct subtypes remains unclear. Using mouse genetics, we reveal that cell of origin is an important determinant of PDAC molecular subtype. Deciphering the biology underlying pancreatic cancer subtypes may reveal meaningful distinctions that could improve clinical intervention. This article is highlighted in the In This Issue feature, p. 521
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 24_Supplement ( 2019-12-15), p. A15-A15
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 24_Supplement ( 2019-12-15), p. A15-A15
    Abstract: The tumor suppressor gene TP53 encodes a transcriptional activator critical for suppressing pancreatic ductal adenocarcinoma (PDAC). TP53 mutations, which are most frequently missense mutations in the DNA binding domain, result in both loss of wild-type p53 function and gain of novel, oncogenic functions. Mouse models of PDAC relying on oncogenic Kras and p53 mutation have suggested that either pancreatic acinar or ductal cells, the two major epithelial cell types in the pancreas, may be the cell of origin for PDAC, although there is very limited understanding of the molecular pathways through which p53 mutations contribute to PDAC driven by oncogenic Kras. Moreover, the cellular functions of p53 involved in tumor suppression remain enigmatic. To set up a platform for deconstructing p53 loss-of-function (LOF) and gain-of-function (GOF) pathways, we have generated mouse models to study p53 function in adult mice, mimicking the context of human cancer, using tamoxifen-inducible Cre recombinases to activate Kras (KrasLSL-G12D) in adult mouse pancreatic acinar (Ptf1aCreER) and ductal (Sox9CreER) compartments, in the backdrop of different p53 alleles. To investigate p53 LOF, we generated mouse cohorts with wild-type p53 expression or conditional p53 knockout (p53fl/fl). To examine p53 GOF, we generated mouse cohorts with conditional expression of structural mutant p53R172H (p53LSL-R172H/fl), contact mutant p53R270H (p53LSL-R270H/fl) and conditional p53 knockout (p53null/fl) for comparison in a uniform p53 heterozygous genetic background of stromal tissues. Our tumor studies have demonstrated that oncogenic Kras expression and p53 knockout or mutation in acinar cells of adult mice leads to metastatic PDAC development with complete penetrance. This suggests a novel role for p53 in tumor suppression by blocking cellular reprogramming of acinar cells to ductal-like cells. In contrast, these mutations drive PDAC from ductal cells less efficiently. To understand the origins of human PDAC, we are investigating the molecular pathways altered in mouse acinar and ductal cell-derived tumors. Through comparison with human pancreatic cancer data, our studies will elucidate the cellular origins of different molecular subtypes of human PDAC. Elucidating the mechanisms of PDAC development is critical to improving detection and therapy in PDAC patients. Citation Format: Brittany M. Flowers, Hang Xu, Kathryn Hanson, Christina Curtis, Hannes Vogel, Laura D. Wood, Laura D. Attardi. Elucidating the role of p53 in the cellular origins of pancreatic cancer development [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2019 Sept 6-9; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2019;79(24 Suppl):Abstract nr A15.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    Proceedings of the National Academy of Sciences ; 2023
    In:  Proceedings of the National Academy of Sciences Vol. 120, No. 10 ( 2023-03-07)
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 120, No. 10 ( 2023-03-07)
    Abstract: The vast majority of human pancreatic ductal adenocarcinomas (PDACs) harbor TP53 mutations, underscoring p53’s critical role in PDAC suppression. PDAC can arise when pancreatic acinar cells undergo acinar-to-ductal metaplasia (ADM), giving rise to premalignant pancreatic intraepithelial neoplasias (PanINs), which finally progress to PDAC. The occurrence of TP53 mutations in late-stage PanINs has led to the idea that p53 acts to suppress malignant transformation of PanINs to PDAC. However, the cellular basis for p53 action during PDAC development has not been explored in detail. Here, we leverage a hyperactive p53 variant—p53 53,54 —which we previously showed is a more robust PDAC suppressor than wild-type p53, to elucidate how p53 acts at the cellular level to dampen PDAC development. Using both inflammation-induced and KRAS G12D -driven PDAC models, we find that p53 53,54 both limits ADM accumulation and suppresses PanIN cell proliferation and does so more effectively than wild-type p53. Moreover, p53 53,54 suppresses KRAS signaling in PanINs and limits effects on the extracellular matrix (ECM) remodeling. While p53 53,54 has highlighted these functions, we find that pancreata in wild-type p53 mice similarly show less ADM, as well as reduced PanIN cell proliferation, KRAS signaling, and ECM remodeling relative to Trp53 -null mice. We find further that p53 enhances chromatin accessibility at sites controlled by acinar cell identity transcription factors. These findings reveal that p53 acts at multiple stages to suppress PDAC, both by limiting metaplastic transformation of acini and by dampening KRAS signaling in PanINs, thus providing key new understanding of p53 function in PDAC.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2023
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 22_Supplement ( 2022-11-15), p. A051-A051
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 22_Supplement ( 2022-11-15), p. A051-A051
    Abstract: Understanding the gene regulatory networks that contribute to PDAC initiation and progression is critical for improving early detection and more effectively treating this disease. Activating mutations in KRAS are often accompanied by mutations in the TP53, CDKN2A, and SMAD4 tumor suppressor genes in ~72%, 30%, and 32% of PDAC cases (Raphael et al. 2017). Understanding the consequences of deletion of individual tumor suppressor genes can delineate specific genotype-phenotype correlations that can provide new clinical insights. Using a genetically engineered mouse PDAC model based on KrasG12D expression and tumor suppressor gene inactivation through Ptf1aCreER expression in adult acinar cells, we showed previously that p53 deletion or point mutation dramatically accelerates PDAC relative to mice with intact p53. To understand tumor evolution in mice with different p53 statuses, we analyzed the transcriptomic profiles of PDACs developing with intact or mutant p53 by spatial transcriptomics and both bulk and single cell RNA-sequencing. These experiments have provided insight into the pathways that become dysregulated as PDAC develops in the context of different p53 alleles. In addition, as oncogenic signals trigger p53 stabilization via the p19ARF (ARF) protein in some contexts, we sought to determine whether ARF acts upstream of p53 in PDAC suppression. Indeed, the frequent deletion of the CDKN2A locus, which encodes both the ARF and p16INK4A (p16) tumor suppressor genes, suggests the importance of both these genes in suppressing PDAC, but the specific role of ARF in PDAC suppression has not been tested. Using mouse models to definitively interrogate the importance of ARF in suppressing PDAC, we found that Arf inactivation promotes KrasG12D-driven PDAC, and with a latency similar to p53 inactivation. Interestingly, concurrent inactivation of both p53 and Arf accelerated oncogenic KRAS-driven PDAC development relative to p53 inactivation alone, suggesting a p53-independent role for ARF in PDAC suppression. Transcriptomic profiling of tumors from these mice revealed both p53-dependent and p53-independent programs of ARF-mediated tumor suppression, which provides insight into how tumors develop in the absence of ARF. Characterization of the immune and fibroblast components of the tumor microenvironment in tumors with intact tumor suppressors, p53 deficiency, Arf deficiency and combined p53;Arf deficiency reveals genotype-dependent differences, including different CAF profiles observed in tumors of different genotypes. Ongoing analysis of human PDAC will help to further parse the roles of ARF and p16 in human pancreatic cancer development. Together, these studies will help to illuminate how p53 and Arf inactivation contribute to cancer cell evolution and crosstalk with the tumor microenvironment during PDAC development, understanding with potential for improving clinical interventions in PDAC. Citation Format: Laura D. Attardi, Brittany M. Flowers, Kathryn Hanson, Abigail S. Mulligan, Sofia Ferreira, Sohinee Bhattacharyya, Hannes Vogel, Laura D. Wood, Mara Sherman. Understanding the Arf-p53 axis in PDAC suppression [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer; 2022 Sep 13-16; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2022;82(22 Suppl):Abstract nr A051.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 3905-3905
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, which is largely due to the intrinsic resistance to a broad spectrum of therapies. Vitamin D (VD) has been extensively studied as a potential preventive or therapeutic agent for PDAC treatment. Although accumulating evidence supports that VD has beneficial effects on PDAC treatment, controversies still exist. The major reason for this inconsistency is that VD resistance widely occurs in PDAC. However, the underlying mechanism by which the VD resistance occurs remains largely unknown. Here, we report that epigenetic primming by glyceryl triacetate (GTA) and 5-azacytidine (5-Aza) overcomes VD resistance in PDAC. Mechanistically, increasing global H3K27 acetylation with GTA and reducing global DNA methylation with 5-Aza not only elevate the VDR expression but also reprogram the VD responsive genes. Consistently, combination treatment with VD analogue calcipotriol (CPT) and epigenetic primming overcomes the VD resistance to inhibit cell proliferation and migration through activating genes involved in negative regulation of cell proliferation and cell migration, while CPT treatment alone has the opposite effects. Intriguingly, further studies in mouse PDAC cells suggest that VDR expression does not always corelate with VD responsiveness. Using the gain and loss of function experiments, we demonstrate that VDR is necessary but not sufficient to trigger the anti-tumor activity of VD, highlighting that epigenetic state is the key determinant of VD responsiveness in PDAC. Collectively, these data reveal a previously undefined mechanism for VD resistance in PDAC and has the potential to guide VD clinical trials in the future. Citation Format: Bo He, Lauren Stoffel, Clifford Jiajun He, Albert Mao Li, Haowen Jiang, Kumsun Cho, Brittany M. Flowers, Sofia Ferreira, Laura D. Attardi, Jiangbin Ye. Epigenetic primming reveals the central role of epigenetic state in the anti-tumor activity of vitamin D in pancreatic ductal adenocarcinoma. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3905.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Research Vol. 81, No. 22_Supplement ( 2021-11-15), p. PO-084-PO-084
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 22_Supplement ( 2021-11-15), p. PO-084-PO-084
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is a very deadly cancer, with a 5-year survival rate of only 9%. This high mortality rate can be attributed to the lack of early detection methods and to tumors being resistant to treatment, in part due to a highly immunosuppressive tumor microenvironment (TME). The most commonly mutated genes in PDAC include activating mutations in KRAS in & gt;90% of cases and mutations in the tumor suppressor TP53 gene in ~72% of cases. However, how p53, which is a transcription factor, normally suppresses PDAC progression is not well understood. Understanding the pathways through which p53 acts to suppress PDAC will provide key insight into the molecular mechanisms underlying PDAC initiation and progression. Our laboratory has recently shown in genetically engineered mouse models that Kras activation, coupled with p53 inactivation, can drive PDAC from either pancreatic acinar or ductal cells, with acinar cell- and ductal cell-derived tumor signatures resembling the classical and basal-like human PDAC subtypes, respectively. To better understand how p53 loss promotes PDAC, we are focusing on the acinar cell-derived PDAC mouse model and using single-cell transcriptomic technologies to ask in which cell states and through which pathways p53 acts to suppress PDAC development, in particular as PDAC has complex interactions between tumor cells and the TME. In this model, PDAC is driven by oncogenic Kras (KrasG12D) along with either p53 wild type (p53wt) or p53 deficiency (p53flox) in adult acinar cells by a tamoxifen-inducible Ptf1aCreER allele. In the progression of this model, acinar cells can give rise to PDAC likely through a transdifferentiation process called acinar-to-ductal metaplasia (ADM), which leads to pancreatic intraepithelial neoplasias (PanINs) and then PDAC. The initial cell fate transition, ADM, can be activated by growth factors, inflammation, and injury. We have conducted single cell RNA-sequencing (scRNA-seq) to compare early stages of tumor development in the presence and absence of p53. We have also conducted spatial transcriptomic analysis of these samples. Using these combined approaches, we are analyzing tumor cell populations and cells of the tumor microenvironment in the p53wt and p53flox genotypes and have found. We will present our latest single cell analyses on p53 dependent cell states and functional pathways during PDAC development. Understanding how tumor cell states and cells of the tumor microenvironment change with p53 status will reveal different paths of PDAC evolution and uncover novel approaches to improve clinical intervention for human PDAC. Citation Format: Kathryn J. Hanson, Brittany M. Flowers, Nicholas Hughes, Hannes Vogel, Le Cong, Laura D. Attardi. The role of p53 in the development of pancreatic ductal adenocarcinoma [abstract]. In: Proceedings of the AACR Virtual Special Conference on Pancreatic Cancer; 2021 Sep 29-30. Philadelphia (PA): AACR; Cancer Res 2021;81(22 Suppl):Abstract nr PO-084.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 10_Supplement ( 2018-05-15), p. A28-A28
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 10_Supplement ( 2018-05-15), p. A28-A28
    Abstract: In the next decade, pancreatic ductal adenocarcinoma (PDAC) is projected to be the second leading cause of cancer-related death. This high rate of mortality is attributed to late-stage diagnosis and lack of effective therapeutic strategies. The tumor-suppressor gene TP53 is mutated in ~75% of PDAC patients. The most frequently observed TP53 mutations in PDAC occur within the DNA binding domain at codons 175 and 273. TP53 mutations may induce loss of wild-type p53 function and gain of novel, oncogenic functions. We aim to use different mouse models of PDAC to identify the molecular pathways deregulated when wild-type p53 is lost and mutant p53 is expressed, to elucidate the molecular mechanisms driving PDAC development. While previous studies have suggested that point mutant p53 drives gain-of-function phenotypes, our studies of aging Pdx1-Cre;KrasLSL-G12D/+;Rosa26LSL-tdTomato/LSL-tdTomato mice with conditional p53 knockout (p53Flox/Flox or p53Flox/Null) or p53 mutation (p53LSL-R172H/Flox or p53LSL-R172H/Null) did not demonstrate a clear mutant p53 gain-of-function phenotype in tumor latency. We hypothesize that tumor latency is too rapid in these models to manifest a clear gain-of-function phenotype, requiring the establishment of different model systems. Indeed, our preliminary studies using tumor-derived p53R172H cell lines and isogenic p53 null cell lines suggest a role for mutant p53 gain of function in later-stage tumorigenesis, specifically in metastatic seeding. Furthermore, we are evaluating the role of the putative PDAC cells of origin, pancreatic acinar and ductal cells, in PDAC development. Using both tumor-derived mouse cell lines and pancreatic lineage-specific genetically engineered mouse models of PDAC, we can evaluate the consequences of loss of wild-type p53 expression and gain of mutant p53 expression in different pancreatic compartments. Ultimately, using these systems we aim to elucidate the target genes and molecular pathways controlled by wild-type p53 or by mutant p53 to regulate PDAC initiation. Understanding the molecular mechanisms driving PDAC initiation is critical for the development of new therapeutic strategies for early detection and effective intervention in PDAC patients. Citation Format: Brittany M. Flowers, Patty B. Garcia, Barbara M. Grüner, Monte M. Winslow, Laura D. Attardi. Understanding the role of the tumor suppressor p53 in pancreatic cancer development [abstract]. In: Proceedings of the AACR Special Conference: Advances in Modeling Cancer in Mice: Technology, Biology, and Beyond; 2017 Sep 24-27; Orlando, Florida. Philadelphia (PA): AACR; Cancer Res 2018;78(10 Suppl):Abstract nr A28.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 22_Supplement ( 2022-11-15), p. A060-A060
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 22_Supplement ( 2022-11-15), p. A060-A060
    Abstract: Understanding the origins of PDAC is critical for developing better clinical interventions. Modeling in mice has revealed that either pancreatic acinar or ductal cells can serve as the cell of origin for PDAC. Using tamoxifen-inducible Ptf1aCreER and Sox9CreER alleles to induce genetic alterations in adult pancreatic acinar and ductal cells, respectively, we showed that oncogenic Kras expression, coupled with p53 inactivation or mutation, can drive PDAC from either pancreatic cell type. PDACs derived from acinar cells resemble the classical subtype of human PDAC, while those derived from ductal cells resemble the basal-like subtype of human PDAC, suggesting that cell of origin is one factor that can influence PDAC subtype. Moreover, the histological lesions preceding cancer are notably different, suggesting that oncogenic events in each cell type result in distinct paths of tumor evolution. Such findings highlight the importance of the earliest events of tumorigenesis in influencing phenotypes and outcomes, and better understanding these different paths of PDAC evolution can therefore be critical for improving early diagnosis and treatment of PDAC. To deepen our understanding of these paths of PDAC development, we have used spatial transcriptomics to characterize the acinar and ductal cell-derived mouse PDACs, to contextualize cell-specific expression programs in cancer cell and stromal cell compartments. The segmentation of transcriptional programs by cell type is of great importance in PDAC due to a characteristically pervasive desmoplastic stroma and low neoplastic cellularity. These studies have validated differences in transcriptomic profiles of the cancer cells in these two types of PDAC, with HIF1 signaling and glycolysis pathway being upregulated in ductal cell-derived PDAC and programs that mirror tissue development and morphogenesis being more prominent in acinar cell-derived PDAC. Our analysis also revealed differences in the fibroblast and immune compartments of acinar and ductal cell-derived tumors, which we are further characterizing, and which will reveal novel strategies for tumor targeting based on the tumor microenvironment. Together, these findings suggest that these two types of PDAC are driven and sustained by distinct pathways and tumor-stromal crosstalk. To help characterize pathways central for cancer cell growth, we also generated cell lines from acinar cell and ductal cell-derived PDACs, and we are characterizing their sensitivity to different agents that modulate pathways specific to each PDAC subtype. Ultimately, this work will help to elucidate how the two major types of human PDAC develop, paving the way in finding subtype-specific vulnerabilities that may be exploited therapeutically. Citation Format: Sofia Ferreira, Brittany M. Flowers, Kathryn J. Hanson, Alberto Gatto, Sohinee Bhattacharyya, Mara H. Sherman, Laura D. Attardi. Characterizing acinar cell and ductal cell derived PDACs in mouse models [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer; 2022 Sep 13-16; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2022;82(22 Suppl):Abstract nr A060.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...