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: Oncotarget, Impact Journals, LLC, Vol. 7, No. 9 ( 2016-03-01), p. 10255-10270
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2016
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Nature, Springer Science and Business Media LLC, Vol. 425, No. 6960 ( 2003-10), p. 851-856
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2003
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Wiley ; 2021
    In:  Journal of Surgical Oncology Vol. 123, No. 3 ( 2021-03), p. 751-759
    In: Journal of Surgical Oncology, Wiley, Vol. 123, No. 3 ( 2021-03), p. 751-759
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) remains a lethal cancer with an urgent need for better medical therapies. Efforts have been made to investigate the efficacy of immunotherapy, particularly given the hallmarks of immune suppression and exhaustion in PDAC tumors. Here, we review the molecular components responsible for the immune‐privileged state in PDAC and provide an overview of the immunotherapeutic strategies for PDAC including vaccine therapy, checkpoint blockade, myeloid‐targeted therapy, and immune agonist therapy.
    Type of Medium: Online Resource
    ISSN: 0022-4790 , 1096-9098
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2021
    detail.hit.zdb_id: 1475314-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2023
    In:  Oncogene Vol. 42, No. 10 ( 2023-03-03), p. 711-724
    In: Oncogene, Springer Science and Business Media LLC, Vol. 42, No. 10 ( 2023-03-03), p. 711-724
    Type of Medium: Online Resource
    ISSN: 0950-9232 , 1476-5594
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2023
    detail.hit.zdb_id: 2008404-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Elsevier BV ; 2013
    In:  Gastroenterology Vol. 144, No. 6 ( 2013-5), p. 1220-1229
    In: Gastroenterology, Elsevier BV, Vol. 144, No. 6 ( 2013-5), p. 1220-1229
    Type of Medium: Online Resource
    ISSN: 0016-5085
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2013
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 22_Supplement ( 2022-11-15), p. PR021-PR021
    Abstract: The tumor microenvironment of pancreatic ductal adenocarcinoma (PDA) includes abundant fibroblasts and infiltrating immune cells, the latter largely immunosuppressive. Immunotherapy approaches have been ineffective in PDA, pointing to the need for a better understanding of the mechanisms of immunosuppression. We previously identified C-C Motif Chemokine Receptor 1 (CCR1) as overexpressed in macrophages exposed to pancreatic cancer cell conditioned medium in vitro. By single-cell RNA sequencing, we found CCR1 to be expressed by tumor associated macrophages (TAMs) and granulocytes in both human and mouse PDA. Conversely, the ligands for CCR1, C-C Motif Chemokine Ligands (CCLs), are produced by tumor associated fibroblasts. Thus, we sought to investigate the functional role of CCR1 in pancreatic cancer using a combination of genetically engineered mouse models and pharmaceutically approaches. In a first set of experiments, we generated KC;CCR1-/- mice (Ptf1a-Cre;LSL-KrasG12D;CCR1-/-) to determine the requirement of CCR1 during oncogenic KRAS induced pancreatic cancer initiation. We did not observe a difference in PanIN formation/progression in KC;CCR1-/- compared to KC mice. However, we observed increased immune infiltration, including CD8 T cells, in KC;CCR1-/- pancreata. In a second set of experiments, we then orthotopically transplanted two independent mouse pancreatic cancer cells derived from the KPC model in syngeneic CCR1 knockout hosts. We observed reduced tumor growth, which was rescued by CD8 T cell depletion, indicating an increase of anti-tumor immunity in mice lacking CCR1. Consistently, we observed elevated cytotoxic Granzyme B expression, as well as an increase of apoptotic cells in tumors harvested from Ccr1-/- mice. Through mass cytometry (CyTOF) and co-immunofluorescence staining we discovered that tumor associated macrophages from CCR1-/- mice expressed less Arginase 1 and CD206, both immunosuppressive markers, compared to macrophages in wild type tumors. In the last set of experiments, we used the CCR1 inhibitor J-113863, administered to mice following establishment of an orthotopically implanted tumor. Similar to the genetic model, CCR1 inhibition resulted in smaller tumors. Further, targeting CCR1 either genetically or using a CCR1 inhibitor synergizes with anti-PDL1 immune checkpoint blockade to reduce tumor growth. Together, our data is consistent with the notion that tumor associated macrophages lacking CCR1 expression are less immunosuppressive, consequently allowing increased CD8 T cell-mediated anti-tumor immunity. Targeting CCR1 in combination with immune checkpoint blockade improves antitumor efficacy in pancreatic cancer. Citation Format: Yaqing Zhang, Wei Yan, Kristee L. Brown, Ashely Velez-Delgado, Zeribe C. Nwosu, Katelyn Donahue, Padma Kadiyala, Sion Yang, Faith R. Avritt, Xi He, Carlos Espinoza, Marina Pasca di Magliano. CCR1 expression defines pancreatic tumor associated macrophages and drives their immunosuppressive properties [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 PR021.
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Molecular Cancer Research Vol. 13, No. 1 ( 2015-01-01), p. 9-15
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 13, No. 1 ( 2015-01-01), p. 9-15
    Abstract: Pancreatic cancer is one of the deadliest human malignancies due to its early metastatic spread and resistance to therapy. The mechanisms regulating pancreatic cancer metastasis are so far poorly understood. Here, using both in vitro and in vivo approaches, it is demonstrated that CD44, a transmembrane glycoprotein expressed on a subset of pancreatic cancer cells, is required for the induction of epithelial–mesenchymal transition (EMT) and the activation of an invasive program in pancreatic cancer. Mechanistically, the transcription factor Snail1 (SNAI1), a regulator of the EMT program, is a downstream target of CD44 in primary pancreatic cancer cells and regulates membrane bound metalloproteinase (MMP14/MT1-MMP) expression. In turn, MT1-MMP expression is required for pancreatic cancer invasion. Thus, these data establish the CD44–Snail–MMP axis as a key regulator of the EMT program and of invasion in pancreatic cancer. Implications: This study sets the stage for CD44 and MT1-MMP as therapeutic targets in pancreatic cancer, for which small molecule or biologic inhibitors are available. Visual Overview: http://mcr.aacrjournals.org/content/early/2014/09/10/1541-7786.MCR-14-0076/F1.large.jpg. Mol Cancer Res; 13(1); 9–15. ©2014 AACR.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 20 ( 2013-10-15), p. 6359-6374
    Abstract: Pancreatic cancer, one of the deadliest human malignancies, is almost invariably associated with the presence of an oncogenic form of Kras. Mice expressing oncogenic Kras in the pancreas recapitulate the stepwise progression of the human disease. The inflammatory cytokine interleukin (IL)-6 is often expressed by multiple cell types within the tumor microenvironment. Here, we show that IL-6 is required for the maintenance and progression of pancreatic cancer precursor lesions. In fact, the lack of IL-6 completely ablates cancer progression even in presence of oncogenic Kras. Mechanistically, we show that IL-6 synergizes with oncogenic Kras to activate the reactive oxygen species detoxification program downstream of the mitogen-activated protein kinase/extracellular signal—regulated kinase (MAPK/ERK) signaling cascade. In addition, IL-6 regulates the inflammatory microenvironment of pancreatic cancer throughout its progression, providing several signals that are essential for carcinogenesis. Thus, IL-6 emerges as a key player at all stages of pancreatic carcinogenesis and a potential therapeutic target. Cancer Res; 73(20); 6359–74. ©2013 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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. C049-C049
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 22_Supplement ( 2022-11-15), p. C049-C049
    Abstract: Pancreatic ductal adenocarcinoma (PDA) is characterized by dense, fibro-inflammatory stroma that is established in early pathogenesis. Cancer-associated fibroblasts (CAFs) make up a significant proportion of the tumor microenvironment and represent a heterogeneous cell population that play both tumor-promoting and tumor-restricting roles. Despite their dramatic expansion in PDA pathogenesis, it is still unclear what cells in the normal pancreas give rise to these CAFs. Lineage-tracing experiments have previously identified pancreatic stellate cells, mesothelial, and tissue-resident fibroblasts as progenitors to subsets of CAFs. In this research, we firstly pulse-labeled cells expressing the mesothelial and fibroblast marker Wilms tumor 1 (WT1) in the normal pancreas of WT1CreERT;R26tdTomato mice and used an orthotopic model of PDA to trace their contribution to the PDA tumor microenvironment. We found WT1+ cells in the normal pancreas expanded to comprise a significant proportion of PDA CAFs according to immunofluorescence. Based upon single-cell RNA-sequencing (scRNA-seq), these WT1+ precursors represent an endogenous pancreatic fibroblast population similar to previously characterized universal adventitial fibroblasts (Pi16+, Dpp4+). To determine the role of WT1 in established PDA, we labeled WT1+ cells following orthotopic tumor implantation into WT1CreERT;R26tdTomato mice, and scRNA-seq analysis revealed that both inflammatory CAFs (iCAFs) and myofibroblastic CAFs (MyCAFs) express WT1. Notably, iCAFs had higher relative expression of WT1+. Additionally, we depleted WT1+ cells with diphtheria toxin from orthotopic tumors established in WT1CreERT;R26tdTomato/iDTR mice. We found depletion of WT1+ cells reduced orthotopic tumor growth, and depleted fibroblasts from the PDA tumor microenvironment. These findings implicate WT1 as a promising lineage tracing marker for a subset of pancreatic fibroblasts and demonstrates a pro-tumorigenic role of WT1+ fibroblasts in PDA. Citation Format: Allison Bischoff, Yaqing Zhang, Wei Yan, Kristee Brown, Wenting Du, Marina Pasca di Magliano. WT1+ cancer-associated fibroblasts promote pancreatic cancer growth [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 C049.
    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 ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 22_Supplement ( 2022-11-15), p. B064-B064
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 22_Supplement ( 2022-11-15), p. B064-B064
    Abstract: Protein phosphatase 2A (PP2A) is a major serine-threonine phosphatase that regulates many cellular pathways including KRAS, whose oncogenic mutation is prevalent in 95% of patients with Pancreatic Ductal Adenocarcinoma (PDAC). Previous research has identified a decrease in global PP2A activity and an increase in the expression of PP2A inhibitors in PDAC cell lines, suggesting that suppression of PP2A activity may be pertinent in PDAC maintenance. Importantly, PP2A has low mutation rates in PDAC, making it a viable target for therapeutic reactivation. While PP2A has been shown to have global tumor suppressive capabilities, the regulation of specific pathways by PP2A can be altered based on PP2A holoenzyme composition. Therefore, there is a critical need to understand the mechanisms by which oncogenic KRAS can affect PP2A function and differential substrate targeting in PDAC. The PP2A holoenzyme consists of 3 subunits: the scaffolding subunit (A), the catalytic subunit (C), and the regulatory subunit (B). There are 16 different B subunits that can be incorporated into the PP2A holoenzyme that are responsible for substrate specificity. The B56α subunit of PP2A has been shown to negatively regulate cellular transformation. Our research aims to investigate the mechanisms by which PP2A-B56α is regulated through oncogenic KRAS and how suppression of B56α impacts the initiation and progression of PDAC. To determine how oncogenic KRAS alters the dynamics of PP2A-B56α and overall PP2A activity we utilized tet-inducible KRASG12D cell lines to allow direct manipulation of KRAS mutational activation. Using this system, we have identified time dependent alterations in cancerous inhibitor of PP2A (CIP2A) following induction of KRASG12D expression, indicating that PP2A suppression may be an early event in PDAC initiation. Consistent with this hypothesis, we characterized changes in the acceleration of PDAC formation in vivo using the Ptf1a-Cre; LSL- KRASG12D (KC) genetic mouse model combined with a B56α hypomorph model (KCBhm/hm). Our data show that the loss of B56α accelerates PDAC initiation, with an increase in pancreatic precursor lesion (PanIN) number and a decrease in healthy acinar area. In response to B56α loss, similar acceleration of acinar to ductal metaplasia (ADM) kinetics were observed in a 3D-cultured ADM Assay. Furthermore, when 3D-cultured acinar cells were treated with a small molecule activator of PP2A (SMAP), SMAP treatment resulted in smaller and fewer ductal structures, preventing the ADM process. Collectively, these data suggest that PP2A-B56α plays a regulatory role in cellular plasticity and loss contributes to PDAC initiation. Future studies will investigate how mutant KRAS-mediated CIP2A expression effects overall PP2A phosphatase activity and how subsequent sequestration of B56α contributes to development of PDAC. Together, these studies identify PP2A as a critical regulator of KRAS-induced cellular plasticity and support reactivation of PP2A as a novel therapeutic strategy in PDAC patients. Citation Format: Samantha L Tinsley, Rebecca A. Shelley, Gagan K. Mall, Ella Rose D. Chianis, Mary C. Thoma, Marina Pasca di Magliano, Goutham Narla, Rosalie C. Sears, Brittany L. Allen-Petersen. The role of PP2A-B56α in KRAS-mediated pancreatic tumorigenesis [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 B064.
    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...