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
Filter
  • American Association for Cancer Research (AACR)  (9)
  • Fiebig, Heinz-Herbert  (9)
Material
Publisher
  • American Association for Cancer Research (AACR)  (9)
Language
Years
Subjects(RVK)
  • 1
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 12, No. 11_Supplement ( 2013-11-01), p. C30-C30
    Abstract: Introduction: MEK 1/2 inhibitors (MEKi) are promising compounds for the treatment of cancer due to frequent activation of the RAS/MAPK/ERK oncogenic pathway. CI-1040 and PD0325901 are newly developed MEKi that are currently being tested in clinical trials. In the present study, we investigated MEKi response in different tumor types and we determined whether an index of KRAS pathway activation (K-PAI) could predict response to MEKi. Material and Methods: CI-1040 and PD0325901 were tested using an ex vivo 3D Tumor Clonogenic Assay (TCA) in a panel of 63 patient-derived tumor xenografts (PDX) covering 15 tumor histotypes. The K-PAI was determined by identifying gene expression patterns (Affymetrix HGU133 plus 2.0 arrays) associated with activation of the pathway and KRAS mutational status (determined by Sanger sequencing). Results: The absolute activities (IC50) of CI-1040 and PD0325901 correlated in most of the tumor models tested (r=0.87). Most of the melanomas were sensitive to both MEKi tested, whereas variable response profiles were observed in colon cancers and non-small cell lung cancers (NSCLC). Ovarian and pancreatic cancer xenografts displayed in most instances weak responses. The KRAS and BRAF statuses were significantly associated with MEKi IC50 (p=0.0001 and p=0.0002, respectively). The melanomas which frequently displayed BRAF mutations (13/21), were highly sensitive to MEKi treatment, whereas ovarian and pancreatic tumors, which frequently harbored KRAS mutations (1/3 and 2/2), were resistant. Moreover, we found that the K-PAI correlated significantly with MEKi IC50 (r & gt;0.5, p & lt;0.0001 for CI-1040 and PD0325901).= Melanomas with low K-PAI values were highly sensitive to MEKi treatment whereas ovarian and pancreatic tumors with high K-PAI values were resistant. Interestingly, the K-PAI was also predictive of response to MEKi treatment for tumors expressing wild-type BRAF and KRAS.Conclusion: This large ex vivo PDX study showed that tumor sensitivity to MEKi is related to histology and to RAS pathway activation. KRAS and BRAF mutations were predictive of MEKi response (resistance and sensitivity, respectively). These results are consistent with published data on cell lines and on small patient cohorts and demonstrate that PDX are adequate models to test targeted drugs such as MEKi. The correlation of tumor sensitivity to MEKi and the RAS pathway activation level (K-PAI) should be further evaluated in ex vivo 3D assays utilizing PDXs or with an in vivo study, to be able to better stratify patients when testing the predictive potential of K-PAI in clinical trials. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):C30. Citation Format: Anne-Lise Peille, Armin Maier, Frederic Foucault, Rebekka Krumbach, Tim Kees, Torsten Giesemann, Thomas Metz, Thomas Metcalfe, Heinz-Herbert Fiebig, Vincent Vuaroqueaux. A KRAS pathway activation index predicting response to MEK inhibitors in patient-derived tumor xenografts. [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 C30.
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 3699-3699
    Abstract: Patients with metastatic V600E mutant melanomas treated with BRAF inhibitors (BRAFi) frequently develop resistant tumors with aggressive phenotypes. Furthermore, approximately 20-40% of V600E mutant tumors are intrinsically resistant to BRAFi. Suitable models for studying mechanisms of acquired and intrinsic resistance to BRAFi are therefore necessary. In the present study, we applied deep sequencing techniques to identify possible mechanisms of intrinsic and acquired resistance in our collection of melanoma patient-derived xenograft models (MEXFs) treated with BRAFi. Mutational and expression profiles in MEXFs were characterized by whole-exome sequencing (WES) and HG-U133 Plus 2.0 Affymetrix chips and correlated with BRAFi efficacy data from 3D Tumor Clonogenic Assays (TCA). In addition, four resistant cell lines were created by continuously treating 2D monolayer cultures of tumor cells with Vemurafenib, all of which were initially sensitive to BRAFi and carry the BRAF V600E mutation. Expression profiles and mutations of these cell lines were analyzed from RNA-seq data. Potential gene candidates responsible for conferring resistance were further investigated by Q-PCR and Western-blot experiments. WES data revealed that the number of mutations per model was highly variable. Some MEXFs showed a hyper-mutated profile ( & gt;2000 mutations) and were characterized by specific mutational signatures in agreement with those found in melanoma (Alexandrov et al, Nature, 2013). Mutation frequencies of genes typically mutated in melanoma are very similar to those found in The Cancer Genome Atlas. We observed a high correlation of mutations between our MEXFs and their respective cell lines. Among the models with a V600E mutation, only one (MEXF 462) showed resistance to BRAFi-treatment as identified by 3D TCA analyses. In this model, we identified gene point mutations and a high over-expression of EGFR, MEK1, PDGFRA and NF1 in contrast to the other models, suggesting a specific regulation of the RTK signaling pathways. In 2D assays, synergistic interaction of Vemurafenib and Erlotinib was shown. RNA-seq analysis of the cell line established from MEXF 276, in which resistance to Vemurafenib was induced, revealed around 20% of genes being differentially expressed (FC & gt;2) between sensitive and resistant cell lines. An up-regulation of EGFR was also found in this resistant cell line and was confirmed by Western-blot. In addition, an overexpression of PLAU, a biomarker of invasiveness, was identified. We currently perform expression profiling of three other models resistant to BRAFi. Preliminary results suggest different patterns of gene regulation involved in acquisition of resistance. A precise map of transcriptomic and mutational profiles of the four cell lines will be generated and we are investigating if invasiveness is increased upon acquisition of resistance. Citation Format: Bruno Zeitouni, Gerhard Kelter, Armin Maier, Florian Kiefer, Frederic Foucault, Anne-Lise Peille, Tim Kees, Torsten Giesemann, Vincent Vuaroqueaux, Thomas Metcalfe, Heinz-Herbert Fiebig. Molecular profiling of BRAFi-resistance in melanoma cancer models using high-throughput sequencing in patient-derived xenografts. [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 3699. doi:10.1158/1538-7445.AM2014-3699
    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 ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 4170-4170
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 4170-4170
    Abstract: Malignant melanomas account for a substantial proportion of cancers worldwide and are largely resistant to conventional therapy. The heterogeneity and the invasiveness of melanomas present notable challenges for anti-cancer treatment. Recently, several mutations in RAF, RAS, and PTEN were identified that allow for prognosis of cancer development and progression. Therefore, inhibitors of the RAF-MEK-ERK pathway form attractive candidates for molecular-based targeted treatment of melanoma cancer. Here, we present the molecular characterization and chemosensitivity profiling of 18 human melanoma cell lines. Eight of these were established by Oncotest from human tumor xenografts originally derived from primary patient material. The molecular profiling included mutational analysis for BRAF, PIK3CA, KRAS, NRAS, TP53, and PTEN by sequencing. The chemosensitivity profiles were determined in vitro using a fluorescence-based cytotoxicity assay and included commonly used chemotherapeutic agents for treatment of melanoma, targeted agents (sorafenib and bortezomib) and various BRAF (PLX-4032, PLX-4720, GDC-0879) and MEK (CI-1040, PD0325901) inhibitors. The clinically relevant BRAF V600E substitution was the most prevalent mutation and was found in more than 50% of the melanoma cell lines. The chemosensitivity profiling revealed diverse patterns of selectivity and potency with mean IC50 values between 0.4 nM (vinblastine) and 8.3 µM (carboplatin). Each of the BRAF inhibitors tested was clearly more active in melanoma cell lines carrying a BRAF mutation. On average, IC50 values of BRAF inhibitors were more than 20-fold lower for the BRAF V600E mutated cell lines compared to the wild type cell lines. However, the BRAF mutated cell line MEXF 462NL was resistant towards BRAF inhibitors and it is warranted to compare its gene expression profile and mutational status with BRAF mutated cell lines responding to BRAF inhibitors. By using an ex vivo clonogenic assay, similar activity patterns for BRAF inhibitors were found, with mutated BRAF tumor xenografts showing markedly higher sensitivity. Potential synergistic activity of BRAF inhibitors with other inhibitors of the RAF-MEK-ERK pathway or with standard chemotherapeutic agents will be presented. The mutational status of selected cancer related genes, as well as responsiveness towards many cytotoxic and targeted agents was analyzed for 18 melanoma cell lines. This extensive genotypic and phenotypic characterization makes this melanoma cell line panel a valuable tool for in vitro profiling of novel compounds, in particular inhibitors of the RAF-MEK-ERK pathway. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4170. doi:1538-7445.AM2012-4170
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 10, No. 11_Supplement ( 2011-11-12), p. A52-A52
    Abstract: Background: PTEN alterations are major determinants of resistance to several new targeted therapies involving EGFR/PI3K/PTEN pathways. Recent findings also suggested implication of its associated PTENP1 pseudogene. PTEN can be altered at the gene or transcriptional level. PTEN expression is often higher in stroma than in tumor cells complicating readouts of assayed tumor samples. Taking advantage of the stroma of xenografted human tumors in nude mice being produced by the host, we characterized specific PTEN/PTENP1 tumor cell transcript expression levels and mutational status in a large panel of tumor models. Materials and Methods: A total of 192 patient-derived xenografts of 23 different histotypes were investigated. Human and murine PTEN, PTENP1 mRNA levels as well as PTEN gene copy numbers were quantified by quantitative polymerase chain reaction using species specific assays; PTEN transcript was analyzed by sequencing. Results were analyzed by tumor type and compared to sensitivity to cetuximab. Results: PTEN transcript alterations were observed in 32 of the 192 tumor xenografts (17%) and were shown to be due to loss of PTEN gene for 9, to loss of transcript expression for 4 and to frameshift or substitution-missense mutations for 14 and 7 samples, respectively. PTEN expression levels and alteration frequency were both associated with specific histotypes. Melanoma and breast cancer showed most PTEN alterations (6/11 and 6/16 respectively). PTENP1 pseudogene was shown to be well expressed in only 61% of tumor xenografts, whereby expression levels depended on the tumor type. No or low PTENP1 expression was found in tumor models having a PTEN alterations. Finally, in a tumor panel consisting of colon, non-small cell lung, gastric and head and neck cancer we found that none of the PTEN altered tumors analyzed were sensitive to cetuximab in vivo. Conclusion: This study confirmed the multiplicity of PTEN alterations occurring in cancer. PTEN expression levels and alterations were associated with PTENP1 expression levels and depended on tumor types. PTEN alterations were shown to be associated with tumor resistance to cetuximab and their impact on other targeted therapies involving EGFR/PI3K/PTEN pathways should be evaluated. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr A52.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3117-3117
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3117-3117
    Abstract: Introduction. Pancreatic cancer (PC) is a devastating disease characterized by high failure rates of the conventional therapies. Increasing evidence suggest that PC resistance to therapies is due to their intrinsic ability to develop high contents of fibrotic stroma. It was recently shown that Hedgehog (HH) pathway can influence PC sensitivity to gemcitabine by modulating stroma production via paracrine mechanisms in between tumor cells and its stroma environment. Taking advantage that the stroma in engrafted human tumors in nude mice is produced by the host, we investigated the differential expression of HH pathway molecules by human PC cells and associated murine stroma in PC xenograft models. For this purpose we developed a new species-discriminating qRT-PCR methodology and analyzed a comprehensive collection of 27 patient-derived PC xenograft models previously characterized for their chemosensitivity. Methods. RNA was extracted from snap frozen PC xenograft samples. After reverse transcription, the samples were assessed using human and mouse species-specific qPCR assays for Tata Binding Protein (TBP), Sonic (SHH), Indian (IHH) and Desert (DHH) Hedgehog ligands, the Hedgehog receptor PATCH1 (PTCH1) and the G-coupled downstream receptor Smoothened (SMO), and for the Zn finger transcription factors GLI1, GLI2, GLI3. Data from human and mouse TBP assays were used for normalization and as surrogate markers of content of human tumor cells vs murine stroma cells in the models. Results. As measured using TBP, the amount of murine stroma in the PC xenograft models is comparable to that in primary PC tumors and could be as high as 50%. The PC xenograft models with the highest amount of stroma were also most resistant to standard therapies. Among the 27 PC xenograft models the expression of the HH molecules was highly variable. However, we consistently evidenced that the human PC cells preferentially express the HH ligands with much lower or neclectable expression of PTCH1, SMO and GLI transcription factors. SHH and at a lower level IHH were found to be predominant compared to DHH. Of note, tumors cells of few selected xenograft models express however a consistent level of HH receptors and GLI transcription factors. On the opposite, the stroma cells were shown to preferentially express the receptors PTCH1 and SMO as well as GLI transcription factors, but not the HH ligands. In nearly all xenograft models, the transcription factor GLI1 and at lower levels GLI2 were more highly expressed than GLI3. Conclusion. In PC xenograft models, HH ligands are expressed by tumor cells, whereas respective receptors and downstream signaling molecules are predominantly expressed in the murine stroma. This result argues in favor of a predominantly paracrine mechanism of HH in most of the PC xenograft models investigated. The activity of HH pathway inhibitors to modulate the tumor stroma and affect tumor growth will be investigated. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3117. doi:10.1158/1538-7445.AM2011-3117
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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. 72, No. 8_Supplement ( 2012-04-15), p. 1725-1725
    Abstract: Introduction: Sorafenib (Nexavar(®)) is a potent multikinase inhibitor suppressing angiogenesis and promoting cell autophagy. Sorafenib has been shown to be active in a broad range of cancers models and is approved in the clinic for treatment of renal cell carcinoma and hepatocellular carcinoma. Sorafenib was shown to inhibit non small cell lung cancers (NSCLC), however a recent phase III study failed to demonstrate efficacy when given together with carboplatin and paclitaxel. Therefore, the identification of molecular determinants of Sorafenib sensitivity is of particular importance for better selecting NSCLC patients for treatment with this therapy. In the present study we used a large collection of NSCLC and renal cancer patient derived tumor models xenografted in nude mice (PDX) to compare Sorafenib anti-tumor activity in both cancer types and to identify molecular determinants of tumor sensitivity. Material and Methods: Sorafenib was tested in vivo in a panel of 33 different PDX including 8 renal and 25 NSCLC. The drug was given orally at 200 mg/kg/day for 12 days and tumor sensitivity was evaluated by tumor volume inhibition of the test group relative to the vehicle control group (T/C in %). Molecular characterization was done using targeted mutation analysis by Sequenom MassARRAY Oncocarta 3.0, Affymetrix SNP V6.0 array for gene copy number variation and HGU133 plus 2 arrays for transcriptome analysis. Results: The mean % T/C values were 42% in renal and 38% in NSCL PDX. Using a T/C value of 30% as cut-off for sensitivity, tumor response to Sorafenib were observed in 5/8 renal (62%) and in 8/25(32%) NSCL PDX. In NSCLC the response to Sorafenib was 2/5 (40%) in epidermoid, 2/7 (28%) in large cell and 3/13 (23%) in adeno subtypes. The transcriptome analysis revealed differentially expressed transcripts between tumors responding to Sorafenib and those being resistant (student t-test adjusted p & lt;0.05). Gene ontology and pathway analyzes will be presented. In addition, for some of these transcripts, differential expression was associated with gene copy number variations in PDX. None of the specific gene mutations detected by Oncocarta panel. 3 were associated with Sorafenib sensitivity. Conclusion: Sorafenib is a potent tumor inhibitor not only in renal but also in the different NSCL cancer model subtypes. Molecular analysis evidenced differentially expressed transcripts but no gene mutation associated with response to treatment. The relevance of these markers for predicting Sorafenib sensitivity should be confirmed in an independent data set. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1725. doi:1538-7445.AM2012-1725
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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. 73, No. 8_Supplement ( 2013-04-15), p. 2774-2774
    Abstract: Introduction: Gastric cancer is common in Asia and Eastern Asia where more than half of the world's cases arise. As is the case for Caucasian patients, amplification and overexpression of the HER2 gene is a critical event in the tumorigenesis of gastric cancer and is present in 10 to 20% of Asian patients. Trastuzumab has been approved for treatment of HER2-positive gastric cancer. However, primary and secondary resistance to Trastuzumab is a significant problem and new strategies to overcome resistance are needed. Models which recapitulate this differential response aid the development of new therapies targeting HER2. We recently enlarged our collection of gastric cancer Patient Derived Xenografts (PDX), which in the past focused on Caucasian patients, by developing PDX from Eastern Asian patients. In this study we investigated whether the newly developed PDX collection is reflective of the clinical situation and contains HER2 amplified/overexpressing tumors. We evaluated the sensitivity to Trastuzumab treatment of gastric cancer PDX which are HER2 amplified/overexpressing and searched for possible additional molecular determinants of sensitivity. Material and Methods: Gastric tumors were xenografted in nude mice and were characterized by Affymetrix SNP V6.0 array and qPCR for gene copy number variation, Sanger sequencing and Sequenom MassARRAY OncoCarta panels 1, 2 and 3 for mutations, Affymetrix HGU133 plus 2.0 arrays for gene expression and using immunohistochemistry (IHC) for protein expression. Response to HER2-targeted therapy was assessed in vivo by treating gastric PDX with Trastuzumab 10 mg/kg/day at days 7, 14, and 21. Results: A total of 29 gastric cancer PDX were established (7 PDX of Caucasian and 22 of Eastern Asian origin). Among these 29 PDX, 1 PDX from a Caucasian patient and 4 from Eastern Asian patients expressed high amounts of HER2 mRNA due to gene amplification that ranged from 10 to & gt;50 copies. IHC in situ analyses revealed that the HER2 amplification/mRNA overexpression correlated with strong HER2 protein expression (score 3+). Ongoing analyses investigating these HER2-amplified/overexpressing PDX for sensitivity to Trastuzumab revealed one PDX sensitive and one PDX resistant to treatment. We will present analyses of key biomarkers such as NRAS/KRAS/BRAF mutations, PIK3CA/PTEN status, HER2 integrity, gene expression profiles and their correlation with sensitivity/resistance to trastuzumab. Conclusion: In agreement with what is observed in clinical practice, we identified Caucasian and Eastern Asian gastric tumors amplified and overexpressing HER2. The PDX from these tumors allowed the investigation of response to therapy targeting HER2. These PDX will aid in testing new HER2 targeted treatments and in identifying potential molecular determinants of resistance to Trastuzumab and other HER2 targeting agents. Citation Format: Vincent Vuaroqueaux, Andreas Ackermann, Jianing Guo, Anne-Lise Peille, Rebekka Krumbach, Frederic Foucault, Thomas Metz, Heinz-Herbert Fiebig. The molecular determinants of sensitivity to HER2 targeted therapy in Patient Derived Xenograft gastric tumor models from Caucasian and Eastern Asian patients. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2774. doi:10.1158/1538-7445.AM2013-2774
    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 ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1203-1203
    Abstract: Cell lines insufficiently recapitulate the genomic structure and diversity of its parent tumor. Consequently, results from in-vitro experiments can often not be reproduced in the clinic. In contrast, patient derived xenograft models (PDX) more closely mimic the molecular characteristics of patient tumors. In the present study, we analyzed a large part of our collection of patient derived xenografts by means of whole-exome sequencing (WES) to investigate whether their genomic alterations were consistent with those observed in patient tumors from “The Cancer Genome Atlas” (TCGA). Exonic regions from 268 Oncotest PDX were targeted using Agilent SureSelect Human all exon kits 38MB or 51MB. Enriched genomic DNA was sequenced with Illumina HiSeq2000 in 100bp paired-end (PE) reads with expected mean coverage of 100X. The WES data was analyzed for concordance with a) Sanger and Sequenom OncoCarta panels b) patient tumor mutational signatures described by Alexandrov et al. (Nature 2013) and c) gene mutation frequencies from publically available data of TCGA tumors. In the present study, we show that most of the mutations previously determined by Sequenom or Sanger sequencing were also detected by WES analyses. The mutations in our models were distributed as follows: 86% non-synonymous SNVs, 8% small indels (1 nucleotide), 3% splicing site and 3% start/stop gains or losses. The number and the type of mutations varied significantly across tumor types. Similar to the findings of Alexandrov et al., who analyzed TCGA tumors, most of our PDX were characterized by [C & gt;T] transitions that correlated with the patient age. High mutation rates were observed in tissues typically exposed to mutagens, such as melanomas (UV radiation) or lung PDX (tobacco smoke), associated with specific mutation signatures such as increased rates of [C & gt;T] or [C & gt;A] mutations, respectively. Furthermore, we found an association between the presence of mutations in DNA repair genes such as MLH1 and increased mutation levels in some tumor types including colon, gastric and pancreatic cancer. Interestingly, in PDX with high mutation levels, frameshift mutations were increased two-fold, and represented 14% of all mutations. Analyzing our PDX gene mutation profiles and those of the TCGA dataset revealed comparable gene alteration patterns across the tumor types, with some exceptions possibly attributed to the enrichment of aggressive tumors in our collection. This study demonstrates that our PDX tumor collection shows typical signatures of mutational processes and gene alteration patterns similar to patient tumors of the TCGA database. These results confirm the relevance of using PDX models to evaluate anticancer agents in preclinical settings. Citation Format: Frederic Foucault, Florian Kiefer, Bruno Zeitouni, Jagatheswari Virayah, Thomas Metcalfe, Vincent Vuaroqueaux, Heinz-Herbert Fiebig. Whole-exome sequencing analysis across 23 histotypes of patient-derived tumor xenografts reveals their similarities with TCGA patient tumors. [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 1203. doi:10.1158/1538-7445.AM2014-1203
    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 ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Molecular Cancer Therapeutics Vol. 10, No. 11_Supplement ( 2011-11-12), p. B177-B177
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 10, No. 11_Supplement ( 2011-11-12), p. B177-B177
    Abstract: Background: Carcinogenesis is characterized by complex genomic rearrangements including gene copy number variations, gene fusions or somatic mutations and may contribute to tumor progression. Increased gene copy number is one of the mechanisms shown to cause over-expression and over-activation of genes and signaling pathways involved in tumor progression. Such alterations offer a rationale to develop drugs specifically blocking these over-expressed genes. Recently, we characterized more than 250 patient-derived tumor xenografts in nude mice on DNA, RNA and protein levels using high throughput technologies. In the present study, we used this database to identify putative target genes that are amplified and over-expressed in our tumor models. Method: A total of 176 patient-derived tumor models representing 26 tumor types have been investigated. DNA was analyzed using Affymetrix genome wide SNP6.0 arrays and RNA transcripts by Affymetrix HGU-133 plus 2.0 arrays. Analysis was done using R-based statistical tools and Pathway Studio. Gene variations were considered relevant when having an increased gene copy number equal to 4 and log of signal intensity from microarray higher than 12. Result: The DNA analysis of the 176 tumor models allowed identification of a total of 2003 different genomic regions with a copy number gain equal to 4. The median number of genomic regions with copy number gains is 15 per tumor model but was shown to greatly vary between tumor types (colon tumors having the lowest median number ∼ 5). The genomic region gains were observed frequently for chromosomes 3, 5, 8, 17 and 18. The size of the 2003 regions with increase copy number gain varied from 100kb to 89mb and included a total of 16.558 protein coding sequences coding. Transcript analysis using Affymetrix data revealed that among these 16.558 coding sequences, 1609 (10%) have a log2 intensity signal above 12 and were considered over-expressed. The pathway studio analysis revealed genes coding for proteins with trans-membrane domain, receptors, kinases and oncogenes. As expected, we identified tumor models with amplified regions containing overexpressed growth factors or receptors such as AREG, EGFR, HER2, c-Met and FGFR1. The analysis also showed new amplified and over-expressed genes associated with cellular growth. These may be putative drug targets. Conclusion: In the present study, we showed that our patient-derived tumor xenograft panel contains representative tumor models with well know amplified and over-expressed genes. The analysis also revealed tumor models with new drug targets genes. The strategies of blocking them at RNA or protein level in appropriate tumor models could contribute to identification of new drugs. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr B177.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    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...