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: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 29, No. 10 ( 2011-04-01), p. 1304-1311
    Abstract: Regional lymph node disease (RLND) is a component of the risk-based treatment stratification in rhabdomyosarcoma (RMS). The purpose of this study was to determine the contribution of RLND to prognosis for patients with RMS. Patients and Methods Patient characteristics and survival outcomes for patients enrolled onto Intergroup Rhabdomyosarcoma Study IV (N = 898, 1991 to 1997) were evaluated among the following three patient groups: nonmetastatic patients with clinical or pathologic negative nodes (N0, 696 patients); patients with clinical or pathologic positive nodes (N1, 125 patients); and patients with a single site of metastatic disease (77 patients). Results Outcomes for patients with nonmetastatic alveolar N0 RMS were significantly better than for patients with N1 RMS (5-year failure-free survival [FFS], 73% v 43%, respectively; 5-year overall survival [OS] , 80% v 46%, respectively; P 〈 .001). Patients with a single site of alveolar metastasis had even worse FFS and OS (23% FFS and OS, P = .01) when compared with patients with N1 RMS; however, the differences was not as large as the differences between patients with N0 RMS and N1 RMS. For embryonal RMS, there was no statistically significant difference in FFS or OS (P = .41 and P = .77, respectively) for patients with N1 versus N0 RMS. Gene array analysis of primary tumor specimens identified that genes associated with the immune system and antigen presentation were significantly increased in N1 versus N0 alveolar RMS. Conclusion RLND alters prognosis for alveolar but not embryonal RMS. For patients with N1 disease and alveolar histology, outcomes were more similar to distant metastatic disease rather than local disease. Current data suggest that more aggressive therapy for patients with alveolar N1 RMS may be warranted.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2011
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Clinical Oncology (ASCO) ; 2010
    In:  Journal of Clinical Oncology Vol. 28, No. 29 ( 2010-10-10), p. e587-e588
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 28, No. 29 ( 2010-10-10), p. e587-e588
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2010
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cell Reports, Elsevier BV, Vol. 24, No. 1 ( 2018-07), p. 238-251
    Type of Medium: Online Resource
    ISSN: 2211-1247
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2018
    detail.hit.zdb_id: 2649101-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5170-5170
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5170-5170
    Abstract: INTRODUCTION: Pediatric rhabdomyosarcoma (RMS) has varying outcomes, particularly in intermediate-risk disease (IR-RMS) due to the limited ability of clinical staging to accurately risk-stratify a large proportion of patients. This study aimed to identify prognostic signatures in IR-RMS, the clinical subgroup with the most heterogeneous outcomes, which can potentially improve risk stratification compared with routine clinicopathologic metrics. Signature performance was validated on an independent set of RMS patients. METHODS: Prospectively obtained primary tumor specimens from 80 IR-RMS patients on Children’s Oncology Group clinical trial protocols formed the training set. Tumors from 54 RMS patients across all clinical risk groups formed the validation set. Whole transcriptome profiling was performed using oligonucleotide microarrays employing nearly 1.4 million probe selection regions (PSRs) and used to derive weighted meta-features. Accuracies of protein-coding and non-coding meta-features to predict overall (OS) and event-free (EFS) survival were compared using areas under receiver operating characteristic curves. Associated biological processes were analyzed using curated pathway analysis tools. RESULTS: PAX-FKHR status was able to predict OS (p=0.041) and EFS (p=0.008) in the validation set, but not in the training set. Histologic subtype followed a similar predictive pattern. Cox regression on over 17,000 coding genes on the training set identified a prognostic 30-coding gene meta-feature (gMF; OS p=0.001, EFS p=0.012). A similar analysis on non-coding transcripts identified a 39-PSR meta-feature (ncMF; OS, EFS p & lt;0.001). Both gMF and ncMF were able to predict OS and EFS (p≤0.023) in the validation cohort. Based on OS, predictive accuracy of ncMF was higher than gMF (96% vs. 71%, p & lt;0.001). Analysis of biological processes using gMF showed enrichment for functions associated with musculoskeletal development and signaling pathways. Similar analysis of non-coding meta-features revealed enrichment for cellular assembly, cell cycle, apoptosis, and cancer-associated functions. CONCLUSIONS: A non-coding RNA meta-feature was able to better predict outcome in IR-RMS than a coding gene meta-feature, where most standard clinical prognosticators failed. The meta-features were independently validated in IR and non-IR RMS. This suggests that non-coding transcripts can regulate and determine RMS biology and aggressiveness, and be used as novel prognostic indicators. Citation Format: Anirban P. Mitra, Sheetal A. Mitra, Jonathan D. Buckley, Stephen X. Skapek, Douglas S. Hawkins, Timothy J. Triche. Coding and non-coding gene meta-features predict outcome in pediatric rhabdomyosarcomas [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5170.
    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 ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 4350-4350
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 4350-4350
    Abstract: BACKGROUND: Features of cancer genomics including gene expression levels can be employed to create biomarker profiles that can predict prior to therapy the diagnosis and prognosis of an individual patient. We aim to translate robust and reproducible diagnostic and prognostic profiles for pediatric solid tumors into clinical tools applicable to all tumor specimens, fresh frozen (FF) or formalin fixed (FFPE). RESULTS: We used Affymetrix GeneChip Human Exon 1.0 ST (HuEx) arrays on 24 cell lines or tumors (4 fusion-positive rhabdomyosarcoma [RMSpos], 5 fusion-negative RMS [RMSneg] , 6 Ewing sarcoma family of tumors [ESFT], 5 neuroblastoma [NB] , and 4 osteosarcoma [OS]) to identify a 41-feature diagnostic metagene that clearly distinguished both the original test samples and a set of validation samples. We then incorporated the 41-feature metagene into a 48-gene panel. To translate this diagnostic signature, we compared the performance of HuEx arrays with several mid-plex assay platforms including Fluidigm's quantitative reverse-transcriptase PCR (q-RT-PCR) and NanoString's nCounter™ Digital Analyzer by measuring gene expression on an 18-cell line panel (4 RMSpos, 4 RMSneg, 5 ESFT, 1 NB, and 4 OS). With q-RT-PCR, we observed r2 values of 0.703 ± 0.085 (RMSpos), 0.558 ± 0.138 (RMSneg), 0.689 ± 0.070 (ESFT), and 0.607 ± 0.121 (OS). nCounter reported r2 values of 0.672 ± 0.171 (RMSpos), 0.620 ± 0.143 (RMSneg), 0.675 ± 0.094 (ESFT), and 0.583 ± 0.129 (OS). Hierarchical clustering with all platforms was able to distinguish the RMSpos, ESFT, and NB cell lines; however, the RMSneg and OS cell lines were less clearly distinguished because some of the OS cell lines lacked high expression of genes characteristic of OS tumors. Cross-platform comparisons yielded good correlation between nCounter and Q-RT-PCR (average r2 = 0.710 ± 0.183). An 18-sample dilution series revealed that EWS-FLI1 type I or PAX3-FKHR transcripts were consistently detected by q-RT-PCR or nCounter even at 1:1000 RNA dilution. Among biological duplicates, q-RT-PCR reported an average r2 = 0.916 ± 0.039 while nCounter obtained average r2 = 0.983 ± 0.011 including raw cell lysates. In addition, we also compared Fluidigm's q-RT-PCR with Applied Biosystems’ Taqman Low-Density Arrays (TLDA) on five NB patient bone marrow samples to detect residual tumor cells. We observed an average r2 = 0.929 ± 0.119 across five NB genes and one housekeeping gene. Finally, we have also successfully profiled FF vs. FFPE tumors on HuEx and are currently applying these mid-plex technologies to paired FF and FFPE samples in order to create diagnostic profiles applicable to routine FFPE material. CONCLUSIONS: Mid-plex platform can reliably distinguish bone and soft tissue sarcoma cell lines and can be used to translate the application of diagnostic signatures. These data warrant further studies to analyze FFPE samples, to compare additional mid-plex platforms, and to test potential prognostic signatures. 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 4350. doi:10.1158/1538-7445.AM2011-4350
    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: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 21, No. 20 ( 2015-10-15), p. 4733-4739
    Abstract: Purpose: Pediatric rhabdomyosarcoma (RMS) has two common histologic subtypes: embryonal (ERMS) and alveolar (ARMS). PAX–FOXO1 fusion gene status is a more reliable prognostic marker than alveolar histology, whereas fusion gene–negative (FN) ARMS patients are clinically similar to ERMS patients. A five-gene expression signature (MG5) previously identified two diverse risk groups within the fusion gene–negative RMS (FN-RMS) patients, but this has not been independently validated. The goal of this study was to test whether expression of the MG5 metagene, measured using a technical platform that can be applied to routine pathology material, would correlate with outcome in a new cohort of patients with FN-RMS. Experimental Design: Cases were taken from the Children's Oncology Group (COG) D9803 study of children with intermediate-risk RMS, and gene expression profiling for the MG5 genes was performed using the nCounter assay. The MG5 score was correlated with clinical and pathologic characteristics as well as overall and event-free survival. Results: MG5 standardized score showed no significant association with any of the available clinicopathologic variables. The MG5 signature score showed a significant correlation with overall (N = 57; HR, 7.3; 95% CI, 1.9–27.0; P = 0.003) and failure-free survival (N = 57; HR, 6.1; 95% CI, 1.9–19.7; P = 0.002). Conclusions: This represents the first, validated molecular prognostic signature for children with FN-RMS who otherwise have intermediate-risk disease. The capacity to measure the expression of a small number of genes in routine pathology material and apply a simple mathematical formula to calculate the MG5 metagene score provides a clear path toward better risk stratification in future prospective clinical trials. Clin Cancer Res; 21(20); 4733–9. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    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. 4730-4730
    Abstract: INTRODUCTION: Pediatric rhabdomyosarcoma (RMS) has varying outcomes, especially in intermediate-risk disease (IR-RMS) due to the inherent inability of clinical staging to accurately risk-stratify a large proportion of patients. This study aimed to identify prognostic signatures in IR-RMS, the clinical subgroup with the most heterogeneous outcomes, which can potentially provide better risk stratification than routine clinicopathologic parameters. Signature performance was validated on an independent set of RMS patients. METHODS: Prospectively-obtained primary tumors from 80 IR-RMS patients on Children's Oncology Group clinical trial protocols formed the training set. Tumors from 19, 15 and 20 patients with low-risk, high-risk and IR-RMS formed the validation set. Annotated and unannotated transcripts were profiled by Affymetrix Human Exon microarrays employing 1,393,765 probe selection regions (PSRs) and used to derive weighted signatures. Potentials of coding and non-coding signatures to predict survival were compared using areas under receiver operating characteristic curves that provided a measure of predictive accuracy. Associated biological processes were analyzed using curated pathway analysis tools. RESULTS: Histologic subtype (p=0.94) and PAX-FKHR fusion status (p=0.66) were unable to predict survival in the training set of IR-RMS. Tumor site was the only clinical predictor of outcome in this set (p=0.041). Cox regression on 17,045 coding transcripts identified a prognostic 30-gene meta-feature (30gMF, p=0.001). Analysis of unannotated transcripts identified a 39-PSR meta-feature (39ncMF) that also predicted survival (p & lt;0.001). Multiple PSRs interrogating the same genomic locus were then replaced by a single PSR that reduced ncMF size to 34 PSRs (34ncMF), which could still predict outcome (p & lt;0.001). Predictive accuracy of 39ncMF was higher than 34gMF (96.4% vs. 70.8%, p & lt;0.001). However, predictive accuracy of the former was comparable to the 34ncMF (96.7%, p=0.54). When applied to the validation set, the 34gMF, 39ncMF and 34ncMF were able to predict outcomes (p=0.022, 0.006, 0.012, respectively). Analysis of biological processes using 34gMF showed enrichment for functions/disorders associated with musculoskeletal development and signaling pathways. Similar analysis of non-coding signatures revealed enrichment for cellular assembly, cell cycle, apoptosis and cancer-associated functions. CONCLUSIONS: A concise non-coding RNA meta-feature was able to better predict outcome in IR-RMS than a coding gene meta-feature, where most standard clinical prognosticators failed. The meta-features were independently validated in IR and non-IR RMS. This suggests that non-coding transcripts can regulate and determine RMS biology and aggressiveness, and be used as novel prognostic indicators. Citation Format: Anirban P. Mitra, Sheetal A. Mitra, Jonathan D. Buckley, Philipp Kapranov, James R. Anderson, Stephen X. Skapek, Douglas S. Hawkins, Timothy J. Triche. Identification of novel prognostic signatures in rhabdomyosarcoma by whole transcriptome expression profiling: A discovery and validation study. [abstract]. In: Pr oceedings 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 4730. doi:10.1158/1538-7445.AM2014-4730
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 2_Supplement ( 2012-01-08), p. A23-A23
    Abstract: Introduction: Rhabdomyosarcoma (RMS) is the most common pediatric soft-tissue sarcoma, stratified by the Children's Oncology Group (COG) into low/intermediate/high risk based on clinical outcomes. However, most patients are categorized as intermediate-risk where survival is highly heterogeneous, thus suggesting an inability to accurately stratify a majority of patients. We profiled intermediate-risk RMS's for levels of coding and non-coding transcripts to construct prognostic signatures. The goal was to identify panels of RNAs that reflect underlying tumor biology and provide better risk stratification than routine clinicopathologic parameters. Methods: Transcriptomes from 79 prospectively-obtained primary tumors from intermediate-risk RMS patients under COG clinical trial protocols were profiled on Affymetrix Human Exon 1.0 ST microarrays. Expressions of 1,400,033 probe sets representing annotated and unannotated transcripts were analyzed using Genetrix suite of microarray analysis tools. Cox regression and leave-n-out cross validation were used to derive and finalize the expression signatures. An effort was made compare individual prognostic potentials of the coding and non-coding signatures, and that of a signature that combined both features. Results: Standard pathologic prognosticators such as histologic subtype classification (alveolar versus embryonal) and PAX-FKHR fusion gene status were unable to predict outcome in this cohort (p=0.40 and 0.45, respectively). Cox regression analysis on 17,049 coding transcripts created a 42-gene meta-feature that was able to predict survival (p=0.00024). Leave-n-out cross validation of this meta-feature upheld its prognostic ability (p=0.00030). Analysis of probe set regions (PSRs) corresponding to unannotated “dark matter” transcripts identified a 32-PSR meta-feature that also predicted survival with greater significance than PSRs corresponding to coding transcripts (p & lt;0.00001). To reduce feature redundancy, multiple PSRs interrogating the same genomic locus were replaced by a representative PSR that shrunk the meta-feature size to 24 PSRs, which was still able to predict survival better than the coding gene meta-feature (p & lt;0.00001). A meta-feature that combined coding and non-coding RNA features retained its ability to predict outcome (p=0.00002), with non-coding RNA features contributing towards the bulk of its prognostic potential. Conclusions: A more concise non-coding RNA meta-feature was able to better predict outcome than a larger coding gene meta-feature in intermediate-risk RMS, where standard pathologic prognosticators failed. This suggests the role of non-coding transcripts in regulating and determining RMS biology and aggressiveness, and their potential to serve as novel prognostic indicators. Citation Format: Anirban P. Mitra, Jonathan D. Buckley, Sheetal A. Mitra, Elai Davicioni, James R. Anderson, Philipp Kapranov, Stephen X. Skapek, Douglas S. Hawkins, Timothy J. Triche. A non-coding RNA panel predicts intermediate-risk childhood rhabdomyosarcoma prognosis better than standard pathologic criteria and coding genes [abstract]. In: Proceedings of the AACR Special Conference on Noncoding RNAs and Cancer; 2012 Jan 8-11; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Res 2012;72(2 Suppl):Abstract nr A23.
    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 ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 20_Supplement ( 2014-10-15), p. A16-A16
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 20_Supplement ( 2014-10-15), p. A16-A16
    Abstract: Introduction: Pediatric rhabdomyosarcoma (RMS) has varying outcomes, especially in patients with intermediate-risk disease (IR-RMS), due to the inherent inability of clinical staging to accurately risk-stratify a large proportion of patients. This study aimed to identify prognostic signatures in IR-RMS patients, the clinical subgroup with the most heterogeneous outcomes, which reflect underlying tumor biology and provide better risk stratification than routine clinicopathologic parameters. Signature performance was further validated on an independent set of RMS patients. Methods: Prospectively-obtained primary tumors from 80 IR-RMS patients on Children's Oncology Group clinical trial protocols formed the training set. Tumors from 19, 15 and 20 patients with low-risk, high-risk and IR-RMS formed the validation set. All patients underwent whole transcriptome expression profiling using Affymetrix Human Exon microarrays. Expressions of nearly 1.4 million probe selection regions (PSRs) representing annotated and unannotated transcripts were analyzed. Cox regression and leave-n-out cross validation were used to derive and finalize the weighted signatures. Potentials of the coding and non-coding signatures to predict overall survival were compared using areas under receiver operating characteristic curves that provided a measure of predictive accuracy. Associated biological processes were analyzed using curated pathway analysis tools. Results: Standard pathologic prognosticators such as histologic subtype and PAX-FKHR fusion status were unable to predict survival in the subset of IR-RMS that comprised the training set (p=0.94 and 0.66, respectively). Tumor site was the only clinical predictor of outcome in the training set (p=0.041). Iterative Cox regression on over 17,000 coding transcripts identified a 30-gene meta-feature (30gMF) that was able to predict survival in the training set (p=0.001). Analysis of PSRs corresponding to unannotated transcripts identified a 39-PSR meta-feature (39ncMF) that also predicted survival in the training set (p & lt;0.001). To eliminate feature redundancy, multiple PSRs interrogating the same unannotated genomic locus were replaced by a representative PSR that reduced the non-coding meta-feature size to 34 PSRs (34ncMF), which was still able to predict outcome (p & lt;0.001). Predictive accuracy of 39ncMF was significantly higher than 34gMF (96.4% vs. 70.8%, p & lt;0.001). However, predictive accuracy of the former was comparable to the non-redundant 34ncMF (96.7%, p=0.54). When the locked signatures were applied to the validation set, the 34gMF, 39ncMF and abbreviated 34ncMF were able to significantly predict outcomes (p=0.022, 0.006, 0.012, respectively). Analysis of biological processes using the coding 34gMF signature showed enrichment for functions associated with skeletal and muscular development and associated disorders, and over-representation of pathways associated with calcium and actin cytokeleton signaling in skeletal muscles. Similar analysis of non-coding signatures revealed enrichment for cellular assembly, cell cycle, apoptosis and cancer-associated functions and p53 signaling. Conclusions: A concise non-coding RNA meta-feature was able to better predict outcome in IR-RMS than a coding gene meta-feature, where most standard clinical prognosticators failed. The prognostic value of these meta-features was independently validated in patients with IR and non-IR RMS. These observations point to the possible role of non-coding transcripts in regulating and determining RMS biology and aggressiveness, and their potential to serve as novel prognostic indicators. Citation Format: Anirban P. Mitra, Sheetal A. Mitra, Jonathan D. Buckley, Philipp Kapranov, James R. Anderson, Stephen X. Skapek, Douglas S. Hawkins, Timothy J. Triche. Discovery and validation of novel prognostic genomic signatures in rhabdomyosarcoma. [abstract]. In: Proceedings of the AACR Special Conference on Pediatric Cancer at the Crossroads: Translating Discovery into Improved Outcomes; Nov 3-6, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;74(20 Suppl):Abstract nr A16.
    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 ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 5581-5581
    Abstract: INTRODUCTION: Rhabdomyosarcoma (RMS), the most common pediatric soft-tissue sarcoma, is stratified by the Children's Oncology Group (COG) into low/intermediate/high risk based on clinical outcomes. Most RMS patients, however, are categorized as intermediate-risk where survival is highly heterogeneous, suggesting an inherent inability to accurately stratify a large proportion of patients. This study profiled intermediate-risk RMSs for expressions of coding and non-coding transcripts with the aim of constructing prognostic signatures. The goal was to identify RNA panels that reflect underlying tumor biology and provide better risk stratification than routine clinicopathologic parameters. METHODS: RNAs extracted from 79 prospectively-obtained primary tumors from intermediate-risk RMS patients under COG clinical trial protocols were profiled on Affymetrix Human Exon 1.0 ST microarrays. Expressions of 1,400,033 probe set regions (PSRs) representing annotated and unannotated transcripts were analyzed using the Genetrix suite of microarray analysis tools. Cox regression and leave-n-out cross validation were used to derive and finalize the expression signatures. Individual prognostic potentials of the coding and non-coding signatures, and that of a signature that combined both features were compared against each other. RESULTS: Standard pathologic prognosticators such as histologic subtype classification (alveolar versus embryonal) and PAX-FKHR fusion gene status were unable to predict outcome in this cohort (p=0.40 and 0.45, respectively). Cox regression analysis on 17,049 coding transcripts created a 42-gene meta-feature that was able to predict survival (p=0.00024). Leave-n-out cross validation of this meta-feature upheld its prognostic ability (p=0.00030). Analysis of PSRs corresponding to unannotated transcripts identified a 32-PSR meta-feature that also predicted survival with greater significance than PSRs corresponding to coding transcripts (p & lt;0.00001). To eliminate feature redundancy, multiple PSRs interrogating the same unannotated genomic locus were replaced by a representative PSR that reduced the meta-feature size to 24 PSRs, which was still able to predict survival better than the coding gene meta-feature (p & lt;0.00001). A meta-feature that combined coding and non-coding RNA features retained its ability to predict outcome (p=0.00002), with non-coding RNA features contributing towards the bulk of its prognostic potential. CONCLUSIONS: A more concise non-coding RNA meta-feature was able to better predict outcome in intermediate-risk RMS than a larger coding gene meta-feature, where standard pathologic prognosticators failed. These observations point to the possible role of non-coding transcripts in regulating and determining RMS biology and aggressiveness, and their potential to serve as novel prognostic indicators. 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 5581. doi:1538-7445.AM2012-5581
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...