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: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 12, No. 11_Supplement ( 2013-11-01), p. B12-B12
    Abstract: Development of resistance is a significant clinical problem for virtually all targeted cancer therapies. We have generated a reproducible, patient derived xenograft (PDX) model of acquired vemurafenib resistance to address these challenges. Continuous treatment of V600E melanoma tumors, caused synchronous tumor stasis for approximately 7 weeks, following which, all tumors displayed simultaneous resistance marked by rapid tumor growth. Additionally, this model maintains the resistance phenotype upon serial transplantation, providing a platform for testing rational drug selection. The fidelity of the PDX models was further confirmed using a BRAF V600V tumor which did not respond to vemurafenib. Onset of vemurafenib resistance is accompanied by increased phosphor-ERK signifying re-engagement of the MAPK signaling pathway and supporting MEK as a potential target. MEK inhibition in vemurafenib resistant tumors using PD0325901, resulted in rapid tumor shrinkage and dramatically reduced phosphor-ERK levels. Treatment of resistant tumors with PD0325901 alone, whilst leading to rapid tumor shrinkage, showed significant host toxicity and onset of acquired MEKi resistance. Interestingly, combination of vemurafenib + PD0325901 was non-toxic, and showed dramatic and sustained tumor suppression. Upon cessation of PD0325901 at 70 days the tumors remained undetectable for the duration of the study ( & gt;100 days). These data support the use of MEK inhibitors post-development of vemurafenib resistance and demonstrate that combination therapy mitigates systemic MEKi toxicity and results in persistent tumor inhibition/eradication. PDX models of acquired resistance provide a unique opportunity to bridge the gap between patients and the basic in vitro biology. Additionally, this PDX system allows the interrogation of the kinetics involved in the development of resistance by longitudinal tumor tissue sampling. Numerous mechanisms have been identified as potential causes of the resistance phenotype. Many have been identified in vitro but not all have been confirmed in patients. We detected no evidence of increased BRAF copy number or expression, although alternative BRAF splicing was identified in resistant tumors. Using differential gene expression accompanied by pathway and network analysis we identified distinct differences in the PDX tumors at various time points during the development of resistance. In particular, a potential role for interferon signaling in resistant tumors was observed. Furthermore, changes in the metabolic profiles were identified with untreated and resistant tumors favoring glycolytic pathways, whereas growth arrested tumors exhibited a preference for oxidative phosphorylation. In conclusion, these results demonstrate the value of PDX models for contributing to clinical cancer management through the decryption of complex drug resistance mechanisms and accelerating the identification of rationally selected drug combinations for bench to bedside applications. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):B12. Citation Format: Noel R. Monks, David J. Monsma, David M. Cherba, Emily Eugster, Dawna Dylewski, Mary E. Winn, Andrew S. Borgman, Paula J. Davidson, Chelsea A. Peterson, Jose M. Pimiento, Alexander E. Ivliev, Yuri Nikolsky, Marina Bessarabova, Valerie S. Calvert, Mariaelena Pierobon, Emanuel F. Petricoin, Craig P. Webb, Brian J. Nickoloff. Overcoming acquired resistance to vemurafenib using clinically relevant PDX models of melanoma. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr B12.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Translational Medicine, Springer Science and Business Media LLC, Vol. 10, No. 1 ( 2012-12)
    Abstract: There is resurgence within drug and biomarker development communities for the use of primary tumorgraft models as improved predictors of patient tumor response to novel therapeutic strategies. Despite perceived advantages over cell line derived xenograft models, there is limited data comparing the genotype and phenotype of tumorgrafts to the donor patient tumor, limiting the determination of molecular relevance of the tumorgraft model. This report directly compares the genomic characteristics of patient tumors and the derived tumorgraft models, including gene expression, and oncogenic mutation status. Methods Fresh tumor tissues from 182 cancer patients were implanted subcutaneously into immune-compromised mice for the development of primary patient tumorgraft models. Histological assessment was performed on both patient tumors and the resulting tumorgraft models. Somatic mutations in key oncogenes and gene expression levels of resulting tumorgrafts were compared to the matched patient tumors using the OncoCarta (Sequenom, San Diego, CA) and human gene microarray (Affymetrix, Santa Clara, CA) platforms respectively. The genomic stability of the established tumorgrafts was assessed across serial in vivo generations in a representative subset of models. The genomes of patient tumors that formed tumorgrafts were compared to those that did not to identify the possible molecular basis to successful engraftment or rejection. Results Fresh tumor tissues from 182 cancer patients were implanted into immune-compromised mice with forty-nine tumorgraft models that have been successfully established, exhibiting strong histological and genomic fidelity to the originating patient tumors. Comparison of the transcriptomes and oncogenic mutations between the tumorgrafts and the matched patient tumors were found to be stable across four tumorgraft generations. Not only did the various tumors retain the differentiation pattern, but supporting stromal elements were preserved. Those genes down-regulated specifically in tumorgrafts were enriched in biological pathways involved in host immune response, consistent with the immune deficiency status of the host. Patient tumors that successfully formed tumorgrafts were enriched for cell signaling, cell cycle, and cytoskeleton pathways and exhibited evidence of reduced immunogenicity. Conclusions The preservation of the patient’s tumor genomic profile and tumor microenvironment supports the view that primary patient tumorgrafts provide a relevant model to support the translation of new therapeutic strategies and personalized medicine approaches in oncology.
    Type of Medium: Online Resource
    ISSN: 1479-5876
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2012
    detail.hit.zdb_id: 2118570-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Pediatric Blood & Cancer, Wiley, Vol. 61, No. 9 ( 2014-09), p. 1570-1577
    Type of Medium: Online Resource
    ISSN: 1545-5009
    Language: English
    Publisher: Wiley
    Publication Date: 2014
    detail.hit.zdb_id: 2130978-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
  • 5
    In: Journal of Translational Medicine, Springer Science and Business Media LLC, Vol. 11, No. 1 ( 2013-12)
    Type of Medium: Online Resource
    ISSN: 1479-5876
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2013
    detail.hit.zdb_id: 2118570-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Molecular Cancer Research Vol. 14, No. 2_Supplement ( 2016-02-01), p. B19-B19
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 14, No. 2_Supplement ( 2016-02-01), p. B19-B19
    Abstract: Triple-negative breast cancer (TNBC) accounts for 15-20% of breast cancers and is associated with advanced stage at diagnosis and poorer outcome compared to other breast cancer subtypes. There is an unmet need for targeted therapeutic strategies for TNBC patients since current treatment options are restricted to standard chemotherapy. Both receptor tyrosine kinase (RTK) and inflammatory signaling have been shown to promote cancer progression and are promising therapeutic targets. Our laboratory was the first to demonstrate that the MET receptor tyrosine kinase is highly expressed in TNBC. Hepatocyte growth factor (HGF), the MET ligand, is highly expressed in breast carcinoma and breast carcinoma-associated fibroblasts (CAFs) and is able to induce paracrine or autocrine MET signaling. MET/HGF signaling is also connected with the pro-inflammatory cytokine interleukin 6 (IL6). HGF and IL6 have been shown to interact to enhance invasion of lung cancer cells and progression of multiple myeloma. In breast cancer patients, high serum expression of HGF and IL6 distinguishes metastatic breast cancers. Nonetheless, there is a gap in knowledge as to whether MET and IL6 signaling pathways directly or indirectly interact and how MET/IL6 activation promotes TNBC progression. We are examining the novel concept that MET and IL6 signaling pathways act through a positive signaling feedback loop to drive TNBC progression. By understanding the interactions between these signaling networks, we will be able to design therapeutic strategies that target critical signaling nodes in TNBC. Analysis of gene expression profiles in the four molecular TNBC subtypes defined by Burstein et al. revealed that MET, HGF, and IL6 are expressed in each of the TNBC subtypes. Immunohistochemical analysis of HGF and IL6 expression in breast cancer tissues revealed significantly higher HGF and IL6 expression in TNBC compared to ER+ breast cancers. To determine the effect of MET and IL6 inhibition, we established Mammary Architecture and Microenvironment Engineering (MAME) 3D co-culture models of TNBC cells ± fibroblasts. In these models, TNBC cells have high MET expression, moderate to high IL6 expression, and minimal IL6 receptor (IL6R) expression; whereas the CAF cells have high HGF expression and moderate IL6R expression. Our preliminary studies revealed that an IL6 neutralizing antibody (siltuximab) reduced TNBC structure volumes relative to IL6 expression in the TNBC cells, whereas an IL6 receptor (IL6R) neutralizing antibody (tocilizumab) had no effect. These results correlate with IL6 and IL6R expression levels in TNBC cell lines. We evaluated the efficacy of MET inhibition using XL184 (cabozantinib) and observed that XL184 significantly inhibited TNBC growth, proliferation, and invasion of diverse TNBC cell lines, yet was ineffective against MET-negative breast cancer cells. We are currently evaluating the effect of HGF-mediated MET activation on IL6 signaling and inhibition in our 3D TNBC models. To evaluate the effect of MET and/or IL6 inhibition in vivo we utilized a novel xenograft mouse model that expresses human HGF (hHGFtg SCID). In TNBC cell lines MDA-MB-231 and HCC70, IL6R inhibition slowed tumor progression marginally, whereas MET inhibition with XL184 and the combination of XL184 + anti-IL6R drastically inhibited tumor growth. In a model of established tumor growth, we started treatment when tumor reached 500 mm3. Again we observed a significant decrease in tumor growth with XL184 treatment (p & lt;0.005), but also observed a decrease in tumor growth with anti-IL6R treatment (p & lt;0.03). In our current studies we are evaluating the effects of XL184 and siltuximab treatment in TNBC + CAF tumors. These studies will identify the RTK and inflammatory signaling networks that are critical for TNBC progression and are potential targets for combination therapy. Citation Format: Elizabeth A. Tovar, Mansoureh Sameni, Curt J. Essenburg, Anita Chalasani, Erik S. Linklater, David M. Cherba, Aruselvi Anbalagan, Mary E. Winn, Bonnie F. Sloane, Carrie R. Graveel. MET and IL6 signaling in triple-negative breast cancer. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Breast Cancer Research; Oct 17-20, 2015; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(2_Suppl):Abstract nr B19.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1213-1213
    Abstract: Background: Most current targeted therapeutics are directed against the activated protein kinase-driven signaling pathway architecture. While guiding therapy selection based on protein phosphorylation is biochemically rational given these proteins represent the direct drug targets it is not certain which protein networks/signaling protein targets are the causal driver of any patient's tumor. Thus, the availability of a system that recapitulates the human in vivo setting and allows for credentialing and identification of which activated signaling networks are causally necessary for tumor viability would be extremely important. Methods: To test two such systems and determine how they resemble the cell signaling architecture of the human primary tumor we compared eight snap-frozen laser capture microdissected (LCM) primary glioblastoma multiforme (GBM) samples with tumor-matching LCM mouse xenografts as well as matching neurospheres (NP). Reverse phase protein microarrays were utilized wherein the total level or phosphorylated level of 70 key signal transduction proteins known to be the direct targets for a number of important targeted therapies and implicated in GBM tumor biology were measured. Results: Overall, the signaling architecture of the xenografts more closely resembled the matched primary LCM tumors in this study set. However, unsupervised hierarchical clustering of the data revealed instances where pathway information such as total c-MET, phospho RAF (S259), phospho GSK3αβ (S21/9) and total EGFR was retained across matched LCM primary tumor, xenografts and NP. In addition there were a number of instances where the signaling architecture of the primary human tumor did not resemble the matched NP and xenograft signature. Conclusion: These pilot data indicate that 3D cell culture conditions may effectively recapitulate in vivo signaling network activation in certain instances, although these preliminary results also suggest that xenografts more closely resemble the cell architecture of matching primary GBM tumors. These data highlight the importance and impact of the microenvironment and culture conditions on tumor cell signal transduction network activation. However, two caveats to this approach remain: (a) drug studies in GBM xenografts and NP are limited to those targeting signaling pathways that are faithfully reproduced from the human in vivo setting and (b) the time required to develop a patient matched xenograft model for individualized drug testing may be too long for a fast-progressing disease such as GBM. Citation Format: Claudius Mueller, Ana C. deCarvalho, Tom Mikkelsen, Laila Poisson, Valerie Calvert, Andrew Borgman, David M. Cherba, Mary E. Winn, Emanuel F. Petricoin. Comparing protein pathway activation mapping portraits between gliobastoma patient-matched primary tumor, xenografts and neurospheres: implications for precision medicine. [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 1213. doi:10.1158/1538-7445.AM2014-1213
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Molecular Cancer Research Vol. 14, No. 2_Supplement ( 2016-02-01), p. B14-B14
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 14, No. 2_Supplement ( 2016-02-01), p. B14-B14
    Abstract: Breast cancers display a remarkable phenotypic diversity that is exploited to promote both tumor progression and therapeutic resistance. Recent studies in several types of cancer have highlighted the significance of intratumoral heterogeneity on both innate and acquired resistance to tyrosine kinase inhibitors (TKIs). Tumor plasticity is supported by the heterogeneous expression of receptor tyrosine kinases (RTKs) and the robustness that the overlapping signaling networks provide. Therefore a thorough understanding of the intratumoral heterogeneity is necessary for the development of effective therapeutic strategies. The receptor tyrosine kinase MET is overexpressed in 20-30% of breast cancers and correlates with poor patient outcome. Previously, we determined that high MET expression correlated with ER-/ERBB2- and basal like breast cancers. These results and the efficacy of MET inhibitors in other cancers suggest that MET may be an effective clinical target for aggressive breast cancer subtypes. Recent studies have exposed interactions between MET and the ERBB receptor family in the progression and therapeutic resistance of several cancers. Since MET, ERBB2, and EGFR are known to be highly expressed in aggressive breast cancer subtypes, it is critical that we understand the relationships between these receptors in order to develop effective treatment strategies. We are investigating the relationship between MET and ERBB receptor signaling in the progression and resistance of ERBB2+ and triple-negative breast cancer (TNBC). We observe that there is a large subset of ERBB2+ breast cancers that express MET and contain MET+/ERBB2+ subpopulations. In a MET+/ERBB2+ breast cancer cell line, MET depletion results in increased ERBB2 activation whereas, ERBB2 depletion results in increased MET activation. Therefore, ERBB2+ breast cancers with MET+ subpopulations may have an innate resistance to ERBB2 inhibition and may benefit from combined MET and ERBB2 inhibition. In TNBC, we observe heterogeneous expression of MET and EGFR. We have developed patient-derived xenografts (PDX) from primary and metastatic TNBCs that have diverse patterns of MET and EGFR expression/activation. In these TNBC PDX models we observe varied responses to monotherapy with MET inhibitors MGCD265 and crizotinib and the EGFR inhibitor erlotinib. Interestingly, therapeutic response to MET inhibition does not correlate with protein expression levels. In all studies, we observe significantly increased efficacy of combination therapy with MET and EGFR inhibition and a decrease in response variability. Examination of phospho-MET localization in treated tumorgrafts revealed that MET and EGFR inhibition induces distinct phospho-MET localization changes. We also observe that in the residual resistant cells phospho-MET is highly expressed in cells with mitotic bodies. We are currently performing phospho-proteomic analysis to determine the effect of MET and/or EGFR inhibition on RTK signaling networks including ERK and AKT pathways. These results will identify proteomic signatures that represent MET and/or EGFR activation/inhibition and sensitivity or resistance to monotherapy or combined MET and EGFR inhibition. Overall these studies give us a more comprehensive view of TNBC network signaling, the level of RTK heterogeneity within TNBC, and the efficacy of MET and/or EGFR inhibition on TNBC progression. Citation Format: Elizabeth A. Tovar, Erik S. Linklater, Curt J. Essenburg, Zach Madaj, David M. Cherba, Mary E. Winn, Hasan Korkaya, Julie L. Boerner, Carrie R. Graveel. Targeting the intratumoral heterogeneity of receptor tyrosine kinases in breast cancer. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Breast Cancer Research; Oct 17-20, 2015; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(2_Suppl):Abstract nr B14.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 22, No. 4 ( 2016-02-15), p. 923-934
    Abstract: Purpose: Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype that is associated with poor clinical outcome. There is a vital need for effective targeted therapeutics for TNBC patients, yet treatment strategies are challenged by the significant intertumoral heterogeneity within the TNBC subtype and its surrounding microenvironment. Receptor tyrosine kinases (RTK) are highly expressed in several TNBC subtypes and are promising therapeutic targets. In this study, we targeted the MET receptor, which is highly expressed across several TNBC subtypes. Experimental Design: Using the small-molecule inhibitor cabozantinib (XL184), we examined the efficacy of MET inhibition in preclinical models that recapitulate human TNBC and its microenvironment. To analyze the dynamic interactions between TNBC cells and fibroblasts over time, we utilized a 3D model referred to as MAME (Mammary Architecture and Microenvironment Engineering) with quantitative image analysis. To investigate cabozantinib inhibition in vivo, we used a novel xenograft model that expresses human HGF and supports paracrine MET signaling. Results: XL184 treatment of MAME cultures of MDA-MB-231 and HCC70 cells (± HGF-expressing fibroblasts) was cytotoxic and significantly reduced multicellular invasive outgrowths, even in cultures with HGF-expressing fibroblasts. Treatment with XL184 had no significant effects on METneg breast cancer cell growth. In vivo assays demonstrated that cabozantinib treatment significantly inhibited TNBC growth and metastasis. Conclusions: Using preclinical TNBC models that recapitulate the breast tumor microenvironment, we demonstrate that cabozantinib inhibition is an effective therapeutic strategy in several TNBC subtypes. Clin Cancer Res; 22(4); 923–34. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Journal of Translational Medicine, Springer Science and Business Media LLC, Vol. 13, No. 1 ( 2015-12)
    Type of Medium: Online Resource
    ISSN: 1479-5876
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2015
    detail.hit.zdb_id: 2118570-0
    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...