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)  (4)
  • 1
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 26, No. 20 ( 2020-10-15), p. 5411-5423
    Abstract: Gene expression–based molecular subtypes of high-grade serous tubo-ovarian cancer (HGSOC), demonstrated across multiple studies, may provide improved stratification for molecularly targeted trials. However, evaluation of clinical utility has been hindered by nonstandardized methods, which are not applicable in a clinical setting. We sought to generate a clinical grade minimal gene set assay for classification of individual tumor specimens into HGSOC subtypes and confirm previously reported subtype-associated features. Experimental Design: Adopting two independent approaches, we derived and internally validated algorithms for subtype prediction using published gene expression data from 1,650 tumors. We applied resulting models to NanoString data on 3,829 HGSOCs from the Ovarian Tumor Tissue Analysis consortium. We further developed, confirmed, and validated a reduced, minimal gene set predictor, with methods suitable for a single-patient setting. Results: Gene expression data were used to derive the predictor of high-grade serous ovarian carcinoma molecular subtype (PrOTYPE) assay. We established a de facto standard as a consensus of two parallel approaches. PrOTYPE subtypes are significantly associated with age, stage, residual disease, tumor-infiltrating lymphocytes, and outcome. The locked-down clinical grade PrOTYPE test includes a model with 55 genes that predicted gene expression subtype with & gt;95% accuracy that was maintained in all analytic and biological validations. Conclusions: We validated the PrOTYPE assay following the Institute of Medicine guidelines for the development of omics-based tests. This fully defined and locked-down clinical grade assay will enable trial design with molecular subtype stratification and allow for objective assessment of the predictive value of HGSOC molecular subtypes in precision medicine applications. See related commentary by McMullen et al., p. 5271
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 11, No. 5 ( 2021-05-01), p. 1082-1099
    Abstract: Effective data sharing is key to accelerating research to improve diagnostic precision, treatment efficacy, and long-term survival in pediatric cancer and other childhood catastrophic diseases. We present St. Jude Cloud (https://www.stjude.cloud), a cloud-based data-sharing ecosystem for accessing, analyzing, and visualizing genomic data from & gt;10,000 pediatric patients with cancer and long-term survivors, and & gt;800 pediatric sickle cell patients. Harmonized genomic data totaling 1.25 petabytes are freely available, including 12,104 whole genomes, 7,697 whole exomes, and 2,202 transcriptomes. The resource is expanding rapidly, with regular data uploads from St. Jude's prospective clinical genomics programs. Three interconnected apps within the ecosystem—Genomics Platform, Pediatric Cancer Knowledgebase, and Visualization Community—enable simultaneously performing advanced data analysis in the cloud and enhancing the Pediatric Cancer knowledgebase. We demonstrate the value of the ecosystem through use cases that classify 135 pediatric cancer subtypes by gene expression profiling and map mutational signatures across 35 pediatric cancer subtypes. Significance: To advance research and treatment of pediatric cancer, we developed St. Jude Cloud, a data-sharing ecosystem for accessing & gt;1.2 petabytes of raw genomic data from & gt;10,000 pediatric patients and survivors, innovative analysis workflows, integrative multiomics visualizations, and a knowledgebase of published data contributed by the global pediatric cancer community. This article is highlighted in the In This Issue feature, p. 995
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 22, No. 2 ( 2016-01-15), p. 301-309
    Abstract: Purpose: This phase II trial evaluated the efficacy and safety of cixutumumab, a human anti–insulin-like growth factor receptor 1 (IGF-1R) monoclonal IgG1 antibody, and explored potential biomarkers in postmenopausal women with hormone receptor–positive breast cancer. Experimental Design: Patients with hormone receptor–positive breast cancer that progressed on antiestrogen therapy received (2:1 randomization) cixutumumab 10 mg/kg and the same antiestrogen (arm A) or cixutumumab alone (arm B) every 2 weeks (q2w). Primary endpoint was progression-free survival (PFS); secondary endpoints included overall survival (OS) and safety. Correlative analyses of IGF-1R, total insulin receptor (IR), and IR isoforms A (IR-A) and B (IR-B) expression in tumor tissue were explored. Results: Ninety-three patients were randomized (arm A, n = 62; arm B, n = 31). Median PFS was 2.0 and 3.1 months for arm A and arm B, respectively. Secondary efficacy measures were similar between the arms. Overall, cixutumumab was well tolerated. IGF-1R expression was not associated with clinical outcomes. Regardless of the treatment, lower IR-A, IR-B, and total IR mRNA expression in tumor tissue was significantly associated with longer PFS [IR-A: HR, 2.62 (P = 0.0062); IR-B: HR, 2.21 (P = 0.0202); and total IR: HR, 2.18 (P = 0.0230)] and OS [IR-A: HR, 2.94 (P = 0.0156); IR-B: HR, 2.69 (P = 0.0245); and total IR: HR, 2.72 (P = 0.0231)] . Conclusions: Cixutumumab (10 mg/kg) with or without antiestrogen q2w had an acceptable safety profile, but no significant clinical efficacy. Patients with low total IR, IR-A, and IR-B mRNA expression levels had significantly longer PFS and OS, independent of the treatment. The prognostic or predictive value of IR as a biomarker for IGF-1R–targeted therapies requires further validation. Clin Cancer Res; 22(2); 301–9. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 4_Supplement ( 2019-02-15), p. P6-04-01-P6-04-01
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 4_Supplement ( 2019-02-15), p. P6-04-01-P6-04-01
    Abstract: BACKGROUND: The most common site of cancer metastases from breast is to bone, which occurs in 65-80% of patients resulting in significant comorbidities caused by pathological fractures, pain, hypercalcemia, and nerve compression. The current standard of care involves surgery, radiation, and treatment with bisphosphonates to target osteoclast driven resorption. These modalities all have limitations, complications, and cannot prevent new bone metastases from developing. There is an obvious unmet need for new therapeutic targets and models to treat and study breast cancer related bone metastasis. A large portion of breast cancer research utilizes cell lines and animal models with tissues taken from early stage, primary breast cancers. Furthermore, endpoints such as tumor size reduction and growth in these studies do not always translate to tumor spread. As a result, these study endpoints are not relevant for individuals that are already living with metastatic disease. MATERIALS and METHODS: Potential therapeutic targets involved in metastasis and osteomimicry were identified by performing exome-capture RNA-sequencing (ecRNA-seq) on eleven matched primary breast tumor and bone metastases. Expression gains and losses were identified from the clinically actionable gene set obtained using the Drug Gene Interaction Database (DGBIdB 2.0). A unique organotypic bone model comprised of endothelial cells (EC), osteoblasts (OB), and mesenchymal stem cells (MSC) co-cultured with primary metastatic bone samples from breast cancer patients is being utilized to target these genes of interest. RESULTS: ecRNA-seq of metastatic bone samples revealed expression gains in genes of interest (GOI): EPH Receptor A3 (EPHA3), Protein Tyrosine Phosphatase, Receptor Type D (PTPRD), Patched 1 (PTCH1), and Platelet Derived Growth Factor Receptor Alpha (PDGFRA). The GOI were highly recurrent in patients with endocrine-resistant disease that had developed bone metastases after treatment but were absent in the de novo bone metastasis cases where patients had not yet received endocrine therapy. Analysis for expression of these GOI as well as osteomimicry genes are being assessed in our organotypic bone model. CONCLUSIONS: The GOI identified in this study have been previously associated with the growth and progression of cancer. Furthermore, these genes may be regulated by the RB1-E2F pathway, which was also found to be upregulated in these same samples, and previous studies have linked this pathway's ability to either directly or indirectly regulate our GOI expression levels. Targeting EPHA3, PTPRD, PTCH1, and PDGFRA in breast cancer recurrences may provide a novel therapeutic approach to treat bone metastases that have developed endocrine resistance in patients. Our organotypic bone scaffold provides a unique model to study the interactions of breast cancer cells with bone cells and to test inhibition of novel target genes. Future development of this model will provide a tool for high-throughput screening of drugs to target bone metastases of breast cancer. Citation Format: Watters RJ, Hersh B, Hankins M, Patel V, Martin B, Li Z, Clark KL, Pirosa A, Priedigkeit N, Weiss KR, Lee AV, Tuan RS, Alexander PG, Oesterreich S. Identification and targeting of clinically actionable genes in bone metastases [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P6-04-01.
    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...