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 Research, American Association for Cancer Research (AACR), Vol. 79, No. 24_Supplement ( 2019-12-15), p. A52-A52
    Abstract: Background: Pancreatic ductal adenocarcinoma (PDA) has a dismal 5-year survival rate of 9%, making this disease one of the deadliest human malignancies. Primary barriers to the treatment of pancreatic cancer include extensive stromal interactions and sustained immune suppression. Aberrant Hedgehog (HH) pathway activity is a hallmark of pancreatic tumorigenesis. Tumor-derived HH ligands signal in a paracrine fashion to the surrounding stroma to influence tumor growth. Expression of HH ligands increases during PDA progression, and previous work has shown that genetic deletion of Sonic HH from the epithelium of mice with pancreatic tumors results in increased Indian HH (Ihh) expression. Methods: Ihh was deleted in tumor cells lines (IhhKO) derived from a genetically engineered mouse model of pancreatic cancer (KrasG12D;Trp;P48-Cre), using CRISPR/Cas-9 gene editing to assess the role of Ihh in the tumor microenvironment. The level of HH signaling was determined using tumor cell co-cultures with Gli1lacZ fibroblasts (derived from mice with a lacZ reporter allele knocked into the Gli1 locus), in which beta galactosidase activity serves as a readout for HH signaling. WT and IhhKO tumor cells were orthotopically transplanted into the pancreas of syngeneic C57BL/6 mice. Human pancreas samples were obtained from surgical resection of pancreatic adenocarcinoma or fine-needle biopsy procedure (FNB). Immune profiling of mouse and human pancreatic tumors was performed using Cytometry Time-of-Flight analysis, and tumor composition was analyzed by single-cell RNA sequencing. In vitro cultures with pancreatic fibroblasts treated with either WT or IhhKO tumor cell conditioned media (CM) were cultured to assess tumor crosstalk. Results: Tumor cells lacking Ihh were generated through CRISPR/Cas-9 deletion, and this was confirmed by qRT-PCR. Co-culture of IhhKO tumor cells with Gli1lacZ fibroblasts results in decreased Gli1 expression both in vitro and in vivo. Immune profiling revealed that tumors lacking Ihh have significantly fewer macrophages (CD11b+/F4/80+), resulting in decreased presence of immunosuppressive factors such as arginase 1 and PDL1. Immune profiling of human PDA revealed similar populations of immunosuppressive myeloid cells present in tumors. In vitro co-cultures demonstrated CCL2 expression was reduced in pancreatic fibroblasts cultured with IhhKO-CM, providing mechanistic insight into the in vivo phenotype observed. Further, scRNA seq analysis suggests that modulation of HH signaling in the tumor microenvironment alters chemokine and immunomodulatory signaling pathways driven by fibroblasts in the pancreatic tumor microenvironment. Significance of Impact: HH signaling in pancreatic fibroblasts contributes to the establishment of an immune-suppressive environment in pancreatic cancer. Combining methods to target HH signaling and immune checkpoint therapy has translational potential in treating pancreatic cancer patients. Citation Format: Nina Steele, Samantha Kemp, Valerie Irizarry-Negron, Veerin Sirihorachai, Ahmed Abbas, Eileen Carpenter, Christopher Halbrook, Carlos Espinoza, Costas Lyssiotis, Howard Crawford, Timothy Frankel, Filip Bednar, Ben Allen, Marina Pasca di Magliano. Modulation of Hedgehog signaling alters immune infiltration in pancreatic cancer [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 A52.
    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 ...
  • 2
    In: American Journal of Gastroenterology, Ovid Technologies (Wolters Kluwer Health), Vol. 116, No. 1 ( 2021-10), p. S35-S35
    Type of Medium: Online Resource
    ISSN: 0002-9270 , 1572-0241
    RVK:
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2021
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Gastroenterology, Elsevier BV, Vol. 162, No. 7 ( 2022-05), p. S-25-
    Type of Medium: Online Resource
    ISSN: 0016-5085
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Gastroenterology, Elsevier BV, Vol. 156, No. 6 ( 2019-05), p. S-373-
    Type of Medium: Online Resource
    ISSN: 0016-5085
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2019
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 22_Supplement ( 2022-11-15), p. PR017-PR017
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is soon to be the second deadliest types of cancer, with a 5-year survival of only 10%. The unique features of PDAC are the expansion of cancer-associated fibroblasts (CAFs), presence of a dense fibrous stroma with high bundled collagen I and immunosuppression, representing a challenge for therapies. Ways to overcome these unique pro-tumor features will be key for the development of better therapeutic strategies for PDAC. Herein we report that, in addition to the identified ectopic expression of NGL-1 in PDAC cells, it is also expressed in the stroma (CAFs and immune cells) of PDAC patients, inversely correlating with overall survival. Stromal NGL-1 was important for tumorigenesis in vivo, as NGL-1 knockout mice (KO) orthotopically allografted with pancreatic cancer cells presented less tumor burden. Further analysis showed that the tumors from KO mice presented more CD8+ T cells and less immunosuppressive cytokines, such as TGFβ. Single cell RNAseq analysis of these tumors showed decreased expression of pro-tumor factors, such as immune checkpoint molecules in T cells and TGFβ related genes across different cellular compartments (epithelial, immune and fibroblasts). In accordance with these results, these tumors presented a limited amount of desmoplastic bundled collagen, suggestive of a TGFβ-deficient environment. In order to further dissect between the NGL-1-dependent contributions of immune vs. local stroma (e.g., CAFs) cells, we generated bone marrow chimeras and performed orthotopic injections to generate tumors. The loss of NGL-1 in each cellular compartment alone failed to phenocopy the full body loss of NGL-1, suggesting that NGL-1 in both immune cells and local stromal are important for tumorigenesis. Functionally, NGL-1 deficient CAFs failed to support the survival of starved PDAC cells in vitro, downregulated important myofibroblastic molecules such as p-smad, and produced less immunosuppressive cytokines, suggesting a role for NGL-1 in key pro-tumor features of CAFs. In fact, the fibroblastic NGL-1 dependent immunomodulatory effects were confirmed with human CD8+ T cells from healthy donors, which lost their cytotoxic profile in the presence of conditioned media (CM) from NGL-1+ CAFs, but were able to keep this profile when exposed to CM from NGL-1 deficient CAFs. Bone marrow derived macrophages from KO mice produced less pro-tumor cytokines and CD8+ T cells lacking NGL-1 proliferated more than those from wild type animals, when stimulated in vitro. Mechanistically, while immune cells and CAFs deficient in NGL-1 are both tumor suppressive, the latter can regain pro-tumor functions in response to TGFβ, explaining the need for a global modulation of NGL-1 expression for an anti-tumor effect. Finally, NGL-1 KO mice orthotopically allografted with PDAC cells responded better (smaller tumors) to chemotherapeutical regimen (FOLFIRINOX) compared to WT animals. All these results point to NGL-1 as a potential new target to modulate immunosuppression and tumorigenesis in pancreatic cancer. Citation Format: Debora Barbosa Vendramini Costa, Ralph Francescone, Janusz Franco-Barraza, Tiffany Luong, Esteban Martinez, Stephen Sykes, Nina Steele, Marina Pasca di Magliano, Dmitry Zhigarev, Charline Ogier, Huamin Wang, Igor Astsaturov, Kerry Campbell, Edna Cukierman. Stromal Netrin G1 ligand (NGL-1): A new modulator of tumorigenesis and immunosuppression in pancreatic cancer [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 PR017.
    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. 79, No. 24_Supplement ( 2019-12-15), p. A54-A54
    Abstract: Pancreatic ductal adenocarcinoma (PDA) has an exceedingly poor prognosis, with only 9% 5-year survival rate. Kras mutations are found in 90% of cases of pancreatic cancer and drive the formation of pancreatic intraepithelial neoplasia (PanIN), precursor lesions to PDA. Both PanIN and PDA are characterized by dense stroma, containing fibroblasts and immune cells. The infiltrating immune cells have a suppressive phenotype and prevent antitumor immunity by cytotoxic T cells. The mechanisms underlying the immunosuppression in pancreatic cancer are only partially understood. Our goal is to investigate the interactions between tumor cells and the immune cells, with the long-term objective to identify new therapeutic targets. Our laboratory has described a mouse model (iKras*) of inducible and reversible expression of oncogenic Kras (Kras*) in the pancreas. Taking advantage of the reversibility of Kras* expression in this model, we conducted a thorough characterization of the immune infiltration and function upon modulation of Kras* at different stages of pancreatic carcinogenesis. iKras* mice and wild-type littermates were enrolled in experiments at the age of 8-12 weeks. Kras* expression was activated, then we induced acute pancreatitis to promote the formation of preneoplastic lesions. After 3 weeks, a time when widespread low-grade lesions and fibrosis are observed, mice were either harvested or Kras* expression was inactivated and 3 days or 1 week later the pancreas was harvested. We performed flow cytometry, immunohistochemistry, and CyTOF in the pancreas to analyze T cell and myeloid cell populations, as well as functional markers (Arg1, iNOS, IFNγ). Myeloid cells and T cells infiltrated the pancreas in presence of active Kras*. Inactivation of Kras* resulted in a relatively modest decrease in infiltrating myeloid cells and a modest increase in CD4+ T cells, particularly regulatory T cells. However, analysis of the functional marker Arg1, a putative immune suppressive molecule expressed in myeloid cells, indicated that its expression depends on Kras*-expressing cells. Kras* in the neoplastic cells initiates the infiltration of immune cells into the pancreas and induces them to express immune-suppressive factors. Going forward, we are investigating the signals downstream of oncogenic Kras that mediate the crosstalk between neoplastic cells and infiltrating immune cells. Citation Format: Ashley Velez-Delgado, Valerie Irizarry-Negron, Rosa Menjivar, Jenny Lazarus, Murali Bollampally, Nina Steele, Timothy Frankel, Fillip Bednar, Yaqing Zhang, Marina Pasca di Magliano. Oncogenic Kras modulates pancreas plasticity and the tumor microenvironment [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 A54.
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 3855-3855
    Abstract: Lung cancer remains the leading cause of cancer-related deaths worldwide, with an estimated 1.6 million deaths each year. Non-small cell lung cancer (NSCLC) is with 85% by far the most common subtype of lung cancer, comprising adenocarcinomas and lung squamous cell carcinoma. Mutations in Kirsten rat sarcoma viral oncogene homolog (KRAS), epidermal growth factor receptor (EGFR) and anaplastic lymphoma receptor tyrosine kinase (ALK) genes are common with the worst overall survival for KRAS mutant adenocarcinoma patients. We have established a murine model of lung cancer, wherein expression of oncogenic Kras can be controlled genetically, allowing activation of oncogenic KrasG12D (Kras*) to initiate tumor growth, tumor eradication upon Kras* depletion and re-activation as a means to model relapse. Oncogenic Kras depletion (deactivation) has previously been reported to result in tumor cell apoptosis even in the presence of tumor suppressor loss. However, the mechanisms of apoptosis, the role of the immune system on these changes, and the mechanisms allowing some tumor cells to escape apoptosis, which typically results in tumor relapse, are unknown. Here, we interrogated the immune response in mediating tumor regression and relapse using this genetically engineered models. Multiplex immunohistochemistry as well as CyTOF provided insight into the changes in immune contexture upon Kras* depletion in mice haploinsufficient for tumor suppressor p53 or mutant for p53 (R172H). Interestingly, total number of T cells including cytotoxic T cells (CTLs) was elevated in lung tumors from p53 mutant mice supporting findings of heightened immune activation and overall response to immune therapy with an increased mutational burden. Kras* inactivation and thus inhibition of MAPK signaling resulted in an overall decrease in abundance of CTLs and antigen presenting cells (APC) as well as engagement of CTL with tumor cells and APCs indicating a decrease in immune presence likely due to proceeding tumor cell kill and immune recruitment. However, intracellular distance of CTL with tumor cells indicated active tumor cell kill of the CTLs to eradicate remaining tumor cells. In summary, these findings support recent observation of increased immune activation in tumors with higher mutational load as well as changes mediated by inhibition of MAPK signaling which both maybe harnessed for enhancing future immunotherapies. Citation Format: Nina Steele, Kristena Y. Abdelmalak, Sarah F. Ferris, Jennifer M. Lee, Carlos Espinoza, Yaqing Zhang, Sundaresh Ram, Craig Galban, Nithya Ramnath, Timothy L. Frankel, Marina Pasca di Magliano, Stefanie Galbán. Oncogenic Kras-mediated regulation of the tumor microenvironment in lung cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3855.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 24_Supplement ( 2019-12-15), p. I04-I04
    Abstract: In human patients and in mouse models, immunosuppressive regulatory T cells (Tregs) accumulate in pancreatic cancer. Tregs are considered a potential therapeutic target with the goal to reverse immunosuppression in a number of malignancies, including pancreatic cancer. Using a genetically engineered mouse model of pancreatic cancer, we determined that—unlike in other malignancies—Treg depletion failed to relieve immunosuppression. Contradicting the current paradigm, depletion of Tregs led to accelerated tumor progression. We show that Tregs are a key source of TGFb ligands both in human samples and in mouse models. Accordingly, their depletion reprograms the fibroblast population, exemplified by loss of smooth muscle actin expression and an increase in the myeloid-recruiting chemokines Ccl3, Ccl6, and Ccl8. Our findings point to a dual role of Tregs in pancreatic cancer, with their immune-suppressive function being balanced by their induction of a SMA-high (tumor-restraining) fibroblast phenotype. Further, our data point to a compensatory immunosuppression mechanism that confers resistance to Treg depletion in pancreatic cancer. Finally, blockade of the common CCL3/6/8 receptor CCR1 combined with Treg-depletion successfully inhibited tumorigenesis, a finding with potential therapeutic implications. Citation Format: Yaqing Zhang, Jenny Lazarus, Nina Steele, Ho-Joon Lee, Wei Yan, Cristopher J. Halbrook, Rosa Menjivar, Samantha B. Kemp, Veerin Sirihoracai, Eileen S. Carpenter, Anna C. Nevison, Alekya Vinta, Michelle A. Anderson, Howard C. Crawford, Costas A. Lyssiotis, Timothy L. Frankel, Filip Bednar, Marina Pasca di Magliano. Regulatory T-cell depletion causes compensatory immune suppression and accelerates pancreatic carcinogenesis [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 I04.
    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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 24_Supplement ( 2019-12-15), p. A62-A62
    Abstract: Objective: Pancreatic cancer is a deadly malignancy, and little progress has been made in the survival of patients during the past 40 years. Harnessing the immune system to elicit anticancer response is an attractive therapeutic option, alone or in combination with antitumor agents. Targeting regulatory T cell (Treg), which acts to suppress immune responses, holds promise as one potential approach to relieve the immune suppression of pancreatic cancer, but functional studies investigating whether Treg depletion at different stages of pancreatic cancer has therapeutic benefit are needed. Methods: We are using KC; Foxp3DTR mice generated by crossing KC (p48-Cre; LSL-KrasG12D) with Foxp3DTR (B6.129(Cg)-Foxp3tm3(DTR/GFP)Ayr/J, Jackson Laboratory) to determine the requirement of Tregs during oncogenic Kras-induced pancreatic cancer precursor lesion formation and progression. We depleted Foxp3-expressing Tregs by Diphtheria Toxin (DT) injection at different time points—either before or after PanIN formation. Multiplex immunohistochemistry and Cytometry by Time-of-Flight (CyTOF) were performed for tumor immunophenotyping; RNA sequencing was used for transcriptomic analysis of the tumor epithelial cells, fibroblasts, and myeloid cells. Results: Surprisingly, instead of blocking tumorigenesis, depletion of Tregs during these early stages led to accelerated tumor progression. We show that Tregs are a key source of TGFb, and accordingly their depletion alters fibroblast population, exemplified by loss of Smooth Muscle Actin expression. Treg depletion further resulted in a compensatory influx of suppressive myeloid cells, thereby preventing an antitumor immune response. Mechanistically, we show that this myeloid recruitment was driven through the CCR1 axis, with the receptor expressed on tumor-associated myeloid cells and several ligands upregulated in epithelial cells and fibroblasts upon Treg depletion. Finally, blockade of CCR1 signaling successfully inhibits tumor progression upon Treg-depletion. Conclusion: Our study revealed an immunosuppressive network of interactions between different components within the pancreatic tumor microenvironment, including Tregs, fibroblasts, tumor cells, and macrophages. Depleting one component (Tregs) can lead to the compensation of immunosuppression from the rest of components within the network. Thus, a thorough understanding of the immunosuppressive network in pancreatic neoplasia is necessary to optimize our approaches to treatment and identify opportunities for combination therapy and prevent potential feedback mechanisms resulting in tumor resistance and relapse. Citation Format: Yaqing Zhang, Jenny Lazarus, Nina Steele, Wei Yan, Ho-Joon Lee, Christopher J. Halbrook, Rosa Menjivar, Samantha B. Kemp, Veerin Sirihorachai, Eileen S. Carpenter, Anna C. Nevison, Alekya Vinta, Michelle A. Anderson, Howard C. Crawford, Costas A. Lyssiotis, Timothy L. Frankel, Filip Bednar, Marina Pasca di Magliano. Regulatory T-cell depletion promotes oncogenic Kras-driven pancreatic carcinogenesis [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 A62.
    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 ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 24_Supplement ( 2019-12-15), p. A25-A25
    Abstract: Pancreatic cancer is a lethal malignancy with a 5-year survival rate of less than 10%. This poor prognosis is, in part, due to patients most often presenting with already metastatic disease. Pancreatic cancer is characterized by an abundant, reactive fibroinflammatory stroma, consisting of cancer-associated fibroblasts (CAFs) and infiltrating immune cells that send and receive signals to and from other nearby cells, including the tumor cells. The crosstalk between tumor cells and elements of the stroma results in an immunosuppressive tumor microenvironment both at the primary tumor and the sites of metastases. A key limitation in studying metastatic disease in mouse models is the inability to predict when the cancer cells will ultimately metastasize. In this study biomaterial scaffolds that mimic the premetastatic niche in vivo, by attracting immune cells followed by tumor cells, were used to longitudinally follow the metastasis process in individual animals. Biomaterial scaffolds were implanted subcutaneously to mimic the premetastatic niche in both syngeneic and spontaneous mouse (iKras* p53*) models of pancreatic cancer. Scaffolds were harvested 21 days post implantation and taken for histologic analysis to characterize the cellular infiltrate in response to the primary tumor. Histologic analysis (H & E, Gomori’s trichrome, and multiplex IHC) shows tumor cell colonization in the scaffold along with a robust immune and fibrotic response. To further characterize the scaffold infiltrate in control versus tumor-bearing mice, mass cytometry (CyTOF) and single-cell RNA sequencing (scRNA seq) analysis was performed. CyTOF analysis revealed the most abundant cell population is heterogenous myeloid cells, which are immunosuppressive in nature. Scaffolds from tumor-bearing animals have an increase in fibroblasts, and subsequently have fewer cytotoxic, CD8+ T cells. scRNA seq analysis of the scaffold infiltrate reveals CD8+ T cells from tumor-bearing animals express higher levels of the immune checkpoint, programmed cell death protein 1 (PD-1) and lower levels of the functional markers, perforin 1 and granzyme B, suggesting the CD8+ T cells are unable to perform their cytotoxic functions. Further analysis revealed a chemokine signaling axis, where fibroblasts recruit distinct subsets of myeloid cells. Taken together, the premetastatic niche (recapitulated by the biomaterial scaffolds) in pancreatic cancer has a high abundance of immunosuppressive myeloid cells, an increase in fibroblasts, and fewer CD8+ T cells, creating an immunosuppressive niche that allows for tumor cell colonization and growth. Future directions include blocking myeloid cell chemotaxis to the scaffold to prevent tumor cell dissemination and reduce metastatic burden. Citation Format: Samantha Kemp, Nina Steele, Eileen Carpenter, Veerin Sirihorachai, Grace Bushnell, Aaron Morris, Carlos Espinoza, Fatima Lima, Zeribe Nwosu, Sophia Orbach, Lonnie Shea, Filip Bednar, Howard Crawford, Marina Pasca di Magliano. Using biomaterial scaffolds to study the genesis of the immunosuppressive premetastatic niche in pancreatic cancer [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 A25.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...