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)
  • 1
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 14, No. 24 ( 2008-12-15), p. 8236-8243
    Abstract: Purpose: Polyamines are important regulators of cell growth and death. The polyamine modulated factor-1 (PMF-1) is involved in polyamine homeostasis. After identifying an enriched CpG island encompassing the PMF1 promoter, we aimed at evaluating the clinical relevance of PMF1 methylation in bladder cancer. Experimental Design: The epigenetic silencing of PMF1 by hypermethylation was tested in bladder cancer cells (n = 11) after azacytidine treatment. PMF1 methylation status was evaluated in 507 bladder tumors and 118 urinary specimens of bladder cancer patients and controls. PMF1 protein expression was analyzed by immunohistochemistry on tissue arrays containing bladder tumors for which PMF1 methylation was assessed (n = 218). Results: PMF1 hypermethylation was associated with gene expression loss, being restored in vitro by a demethylating agent. An initial set of 101 primary frozen bladder tumors served to identify PMF1 hypermethylation in 88.1% of the cases. An independent set of 406 paraffin-embedded tumors also revealed a high PMF1 methylation rate (77.6%). PMF1 methylation was significantly associated with increasing stage (P = 0.025). Immunohistochemical analyses revealed that PMF1 methylation was associated with cytoplasmic PMF1 expression loss (P = 0.032). PMF1 protein expression patterns were significantly associated with stage (P & lt; 0.001), grade (P & lt; 0.001), and poor overall survival using univariate (P & lt; 0.001) and multivariate (P = 0.011) analyses. Moreover, PMF1 methylation in urinary specimens distinguished bladder cancer patients from controls (area under the curve = 0.800). Conclusion: PMF1 was identified to be epigenetically modified in bladder cancer. The association of PMF1 methylation with tumor progression and its diagnostic ability using urinary specimens support including PMF1 assessment for the clinical management of bladder cancer patients.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2008
    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: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 27, No. 6_Supplement ( 2021-03-15), p. S12-04-S12-04
    Abstract: Background: Previous reports indicate that lung cancer patients are at an increased risk of severe COVID-19 disease and higher mortality rate compared to general population. However, prognostic factors are not yet clearly identified. The LunG canceR pAtients coVid19 Disease (GRAVID) study aimed to describe clinical characteristics, outcomes and predictors of poor prognosis in patients with lung cancer and COVID-19. Methods: In this large nationwide prospective study, medical records of lung cancer patients with COVID-19 diagnosis from 65 spanish hospitals were included. Clinical features, treatments and disease outcomes were collected. The primary endpoint was to determine any-cause mortality; secondary endpoints were hospitalization and admission at intensive care units (ICU). Risk factors of poor prognosis were identified by univariable and multivariable logistic regression models. Results: Overal, 447 patients were analysed. Mean age was 67.1 ± 9·8 years, and the majority were men (332, 74·3%) and current/former smokers (383 (85.7%). NSCLC was the most frequent cancer type (377, 84.5%), being adenocarcinoma (228, 51·0%) the predominant histology. 354 patients (79.2%) had unresectable stage III or metastatic disease, and 266 (59.5%) where receiving anticancer treatment, mostly first-line chemotherapy. 350 (78.3%) patients were hospitalized for a mean of 13·4 ± 11·4 days, 9 (2.0%) patients were admitted to ICU, and 146 (32.7%) patients died. Advanced disease and corticosteroid treatment at hospitalization were predictors of mortality. Non-terminal stage hospitalized patients with lymphocytopenia and high LDH showed an increased risk of death. Severity of COVID-19 correlated to mortality, admission at ICU and mechanical ventilation. Conclusion: With underlying comorbidities and immunocompromised status, patients with lung cancer and COVID-19 present high hospitalization and mortality rates. These outcomes, alongside the identification of prognostic factors, may inform physicians on risks and benefits for this population to provide individualized oncological care. Citation Format: Jose Maria Mazarico Gallego, Antonio Calles, Monica Antoñanzas, Cristina Pangua, Xabier Mielgo Rubio, Ernest Nadal, Rafael Lopez Castro, Ana Lopez-Martin, Edel del Barco, Manuel Domine, Virginia Calvo, Pilar Diz, Carmen Sandoval, Elia Sais Girona, Ivana Gabriela Sullivan, M. Angeles Sala, Gema Garcia Ledo, Jose Ramon Jarabo, Rosa Alvarez Alvarez, Javier Baena, Maria Gonzalez-Cao, Mariano Provencio. COVID-19 disease in patients with lung cancer in Spain: GRAVID Lung Cancer Patients Disease (GRAVID study) [abstract]. In: Proceedings of the AACR Virtual Meeting: COVID-19 and Cancer; 2021 Feb 3-5. Philadelphia (PA): AACR; Clin Cancer Res 2021;27(6_Suppl):Abstract nr S12-04.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Clinical Cancer Research Vol. 26, No. 11_Supplement ( 2020-06-01), p. B55-B55
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 26, No. 11_Supplement ( 2020-06-01), p. B55-B55
    Abstract: The PI3K/AKT/mTOR signaling pathway is one of the most frequently mutated pathways in human cancers, including breast cancer. Given the current urge for more personalized approaches, there is an increasing need for a rigorous list of biomarkers and detection methods with advanced, less-invasive, reliable, and low-cost technologies. Despite limited clinical results to date, the detection of PIK3CA mutations shows great potential to contribute in cancer patient management in the future at many different levels, including diagnosis, treatment choice and monitoring, and identification of drug resistance. However, current molecular methods still lack analytical sensitivity to detect mutations of low abundance in high wild-type DNA background. Makrigiorgos et al. recently developed a nuclease-assisted minor-allele enrichment assay with overlapping probes, which is practical and cost-effective and allows detection of very-low-abundance mutations that could have relevance in the clinic. Thus we aimed to develop PIK3CA mutation-specific nuclease-based enrichment assay and validate it with tissue biopsy and blood samples using crystal dPCR as well as combine it with our developed Sybr Green real-time qPCR detection method (Alvarez-Garcia et al. 2018), which allows a quick detection of PIK3CA mutations in clinical samples at low cost. Citation Format: Ieva Keraite, Virginia Alvarez-Garcia, Jurgen Del Favero, Maiwenn Kersaudy-Kerhoas, Nicholas Leslie. PIK3CA mutation enrichment and detection in clinical samples [abstract]. In: Proceedings of the AACR Special Conference on Advances in Liquid Biopsies; Jan 13-16, 2020; Miami, FL. Philadelphia (PA): AACR; Clin Cancer Res 2020;26(11_Suppl):Abstract nr B55.
    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 ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 4_Supplement ( 2020-02-15), p. OT1-07-06-OT1-07-06
    Abstract: Background: The HER3 protein is expressed in multiple solid tumor types, including breast cancer, and high expression of HER3 is associated with poor prognosis. U3-1402 is a novel HER3-targeted antibody-drug conjugate designed with a peptide-based cleavable linker and a topoisomerase I inhibitor payload. U3-1402 has a high drug-to-antibody ratio (~ 8:1), and the stable linker is selectively cleaved by lysosomal enzymes upregulated in tumor cells. In addition, high cell membrane cross-penetration allows for potential payload activity against neighboring tumor cells with antigen heterogeneity. This ongoing phase 1/2, multicenter, open-label, first-in-human study was initiated in Japan and expanded to the United States to evaluate the safety and efficacy of U3-1402 in HER2-negative (including hormone receptor [HR]-positive disease and TNBC), HER3-expressing advanced/unresectable or metastatic breast cancer (NCT02980341/JapicCTI-163401). The study comprises 3 parts: dose escalation, dose finding, and dose expansion. Preliminary results from the phase 1 dose-escalation and dose-finding parts demonstrated the recommended dose for expansion, with antitumor activity of U3-1402 in this heavily pretreated patient population (Masuda, et al. SABCS 2018). Methods: In the ongoing, phase 2, dose-expansion part of this trial, U3-1402 is administered via intravenous infusion every 3 weeks to patients with HER3-expressing advanced/unresectable or metastatic breast cancer. Approximately 110 patients will be treated in the dose-expansion part within cohorts based on breast cancer molecular subtype and HER3 protein expression: patients with HER3-high, HER2-negative, HR-positive disease will receive 4.8 or 6.4 mg/kg U3-1402; patients with HER3-low, HER2-negative, HR-positive disease will receive 6.4 mg/kg U3-1402; and patients with HER3-high, TNBC will receive 6.4 mg/kg U3-1402. Eligible patients for dose expansion are aged ≥ 18 years in the United States (≥ 20 years in Japan); have documented HER3-positive disease, as measured by immunohistochemistry; must undergo fresh tumor biopsy prior to starting treatment, if a sample has not already been submitted for assessment of HER3 expression. For all cohorts except TNBC, patients must have received ≥ 2 and ≤ 6 prior chemotherapy regimens, including ≥ 2 regimens for advanced/unresectable or metastatic disease. For the TNBC cohort, patients must have progressed after receiving 1 to 2 prior chemotherapy regimens in the advanced setting. Patients with previously treated or asymptomatic untreated brain metastases are eligible. Treatment will continue until progression, unacceptable toxicity, death, withdrawal of consent, or termination of the study. The primary objective of dose expansion is to evaluate the safety and efficacy of U3-1402; the secondary objectives are to evaluate the relationship between the efficacy of U3-1402 and HER3 expression and to assess pharmacokinetics and antidrug antibodies. Efficacy assessments include investigator-assessed objective response rate per RECIST v1.1, response duration, time to response, clinical benefit rate, progression-free survival, overall survival, and percent change in target lesion(s). Patients receiving at least 1 dose of U3-1402 with pretreatment and posttreatment tumor assessments will be evaluated for efficacy. Citation Format: Ian Krop, Norikazu Masuda, Takahiro Kogawa, Shunji Takahashi, Kan Yonemori, Kenichi Inoue, Takahiro Nakayama, Yutaka Yamamoto, Ricardo Alvarez, Tatsuya Toyama, Akihiko Osaki, Masato Takahashi, Joyce O'Shaughnessy, Yasuaki Sagara, Shigehira Saji, Virginia Kaklamani, Sun Young Oh, William Gradishar, Barbara Haley, Tsutomu Iwasa, Tiffany Traina, Naoto Ueno, Steve Isakoff, Shoichi Ohwada, Yoshimi Tanaka, Sabeen Mekan, Hiroshi Onuma, Om Sharma, Hiroji Iwata. Phase 1/2 first-in-human study of U3-1402, an anti-human epidermal growth factor receptor 3 (HER3) antibody-drug conjugate, in HER3-expressing advanced/unresectable or metastatic breast cancer, including those with triple negative breast cancer (TNBC) or HER3-low disease [abstract] . In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr OT1-07-06.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 4_Supplement ( 2022-02-15), p. P2-09-06-P2-09-06
    Abstract: Hereditary susceptibility studies for breast cancer are key to enhancing early detection and exercising prevention strategies in order to reduce breast cancer mortality. Germline pathogenic variants that have shown susceptibility to breast and ovarian cancer are detected in gene panels including ATM, BRCA1, BRCA2, BRIP1, CDH1, CDKN2A, CHEK2, DICER1, EPCAM, MLH1, MSH2, MSH6, NBN, NF1, PALB2, PMS2, PTEN, POLE, RAD50, RAD51C, RAD51D, RECQL, STK11, and TP53. Historically, minority populations have been overlooked both in reach and access to genetic testing, resulting in a lack of knowledge of the genomic landscape and creating a barrier to the application of genetics in clinical medicine. This is the case with the populations of South Texas including the Texas-Mexico border region. According to the US Census Bureau, the South Texas region population is comprised of 69% Hispanics while the Texas-Mexico border region population is comprised of over 90% Hispanics. Previous studies have shown multiple pathogenic variants and also variants of unknown significance (VUS) specific to ethnic populations and regions, but there is little information regarding the Mexican American population of South Texas.Our GRACIAS Texas Program has provided services to individuals and professionals across 26 counties in the South Texas area by specifically providing cancer genetic counseling and testing to individuals and families at highest risk. Recruitment was accomplished through comprehensive outreach to communities through health fairs, small group education sessions, mammography centers, hospitals, FQHCs, and direct interactions with medical providers. Although our program focused on recognizing both breast and colon cancer, and also reached those with rare cancer predisposition syndromes, the concern for breast cancer risk was most commonly addressed, accounting for the majority of cases. A total of 1595 individuals were identified as appropriate for cancer genetic counseling, and of these, 1377 individuals completed genetic testing. Of all individuals who received cancer genetic counseling, 1269 (79.5%) were Hispanic, 16% non-Hispanic White, 3% African American, and 1% other race/ethnicity. The group of individuals receiving testing consisted of 86% females and 14% males. Of those tested, 259 (18.8%) individuals were found to have pathogenic genetic variant and 187 (13.7%) individuals were found to have a VUS. Notedly, the VUS were nearly twice as common in the Hispanic population (14%) as compared to non-Hispanic White population (7.2%). This underscores the disparities of knowledge in genomic variation in Hispanic and non-Hispanic population. We also note that although the percentage of African American patients seen was small, 12 of 45 (26.6%) were found to have a VUS which further reflects the underrepresentation of African Americans in genomic landscape studies.These variants are important because they present a dilemma when advising patients as to need for cancer screening. However, recognition and further exploration of these VUS provide a future pathway to functional assessment and thus eventual knowledge to guide patient care. We continue to monitor for any changes to the status of these VUS. Although many variants are ultimately classified as benign variations, we have seen cases in which a VUS is reclassified into a likely pathogenic or definitely pathogenic variation having significant implications for screening, prevention, and management for these individuals and their families emphasizing further the need to continue to follow individuals tested. Our observations will help define the gene-specific risks of individuals and families in our underserved communities and will support the goal of closing gaps in genomic disparities.Supported by CPRIT grants PP120089 and PP160011 and NIH P30 CA54174. Citation Format: Stephanie Soewito, Rachel Wyatt, Emily Berenson, Natalie Poullard, Shawn Gessay, Lindsey Mette, Kristin Shelby, Elise Alvarez, Clarissa Aviles, Anna Maria Pulido Saldivar, Pamela Otto, Ismail Jatoi, Virginia Kaklamani, Gail E Tomlinson. Increased rates of genetic variants of unknown significance in Latino and African American populations of south Texas [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P2-09-06.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 4673-4673
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 4673-4673
    Abstract: BACKGROUND: Urothelial carcinomas presenting T1G3 non muscle-invasive lesions are high-risk tumors because of the high recurrence and progression to muscle invasive disease rates. The Bacillus of Calmette-Guérin (BCG) is a standard treatment to reduce tumor recurrence and delay progression of high-risk non-muscle invasive bladder tumors. However, it is not clear yet which T1G3 patients are more prone to display a more aggressive clinical behavior or be susceptible to respond to BCG. OBJECTIVE: The aim of this study was to evaluate the role of myopodin methylation as a clinical outcome prognosticator and predictive biomarker for BCG response in patients with T1G3 bladder tumors. DESIGN, SETTING, AND PARTICIPANTS: The methylation status of myopodin was analyzed on tumor specimens belonging to 170 patients with T1G3 non-muscle invasive bladder cancer, being a subset of them undergoing BCG treatment (n=108). MEASUREMENTS: Myopodin methylation was assessed by methylation-specific polymerase chain reactions. Recurrence, progression into muscle invasive tumors and disease-specific overall survival rates were analyzed using competing risks regression analysis. RESULTS: Among the 170 cases analyzed, 72 of them recurred (42.4%), 36 progressed (21.2%), and 24 died of the disease (14.1%). Univariate and multivariate survival analyses revealed that myopodin methylation was significantly associated with an increased recurrence rate (p=0.004), progression (p=0.002), and shorter disease-specific overall survival (p=0.020). Interestingly, in a subset of cases treated with BCG, myopodin methylation was also related to an increased recurrence rate (p=0.011), progression (p=0.030), and a shorter disease-specific overall survival (p=0.028). CONCLUSIONS: Epigenetic analyses revealed that myopodin methylation was associated with the clinical outcome of patients with T1G3 tumors. Myopodin methylation was related to tumor aggressiveness in patients with T1G3 disease undergoing BCG treatment. Myopodin methylation distinguished patients responding to BCG from those who may require a more aggressive therapeutic approach. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4673.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    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: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 10, No. 11_Supplement ( 2011-11-12), p. B227-B227
    Abstract: The PIM family of serine/threonine kinases (PIM-1, 2 and 3) has been originally identified as proviral integration sites involved in lymphomagenesis induced by murine leukemia virus. In almost all of the MuLV-induced lymphomas in Eμ-Pim-1 transgenic mice either c-MYC or N-MYC is activated by proviral insertion. The oncogenic properties of Pim kinases were shown to encompass their capacity to counteract the increased sensitivity to apoptosis induction that is associated with MYC-driven tumorigenesis. Pim-1, 2 and 3 are highly conserved kinases that have unique structural properties, and are characterized by a constitutive serine/threonine activity that does not depend on post-translational modifications for activation. Pim kinases activity supports in vitro and in vivo tumor cell growth and survival through modification of an increasing number of common as well as isoform-specific substrates. Pim expression is driven by oncogenes as BCR-ABL, JAK2, FLT3 and KRas. Pim kinases are overexpressed in a range of haematopoietic malignancies and solid cancers, and its overexpression is associated with drug resistance. Inhibition of PIM kinase activity may be an attractive therapeutic strategy with possible favourable toxicity profiles due to minimal phenotype of mice mutant for all Pim family members. Based on the knowledge of complexes of ligands with PIM1 protein, previously reported, as well as known PIM1 inhibitors coming from and internal HTS campaign, we carried out a scaffold hopping strategy for hit generation. We identified triazolopyridine compounds with potent activity against PIM and FLT3 kinases. We have explored different substitution patterns to improve selectivity of PIM vs FLT3 activity. Here, we describe the exploration and biological characterization of this bicyclic series, reporting its SAR/SPR (ADME). We identified lead compounds with potency in the low nanomolar range vs. PIM1, 2 and 3 and high selectivity versus a panel of 24 protein kinases. The compounds display cellular activity by blocking PIM signaling, S112 P-Bad in H1299 Pim1 cells, in the low nanomolar range. The combination of the PI3K inhibitor GDC-0941 with Pim inhibitors was strongly synergistic in vitro in non solid and solid tumoral cell lines. Finally, we assessed the effect of Pim kinase inhibition on Pim signaling and tumor growth in vivo in a mouse intravenous tumor xenograft model employing tumoral cells coming from E myc mice and in an inducible transgenic KRasV12 mouse model. When delivered orally, ETP-995 inhibited phosphorylation of bad and 4EBP and reduced c-myc expression in vivo, and the compound significantly inhibited tumor growth. 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 B227.
    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 ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1240-1240
    Abstract: Hereditary breast and ovarian cancer syndrome (HBOC) is an autosomal dominant disease that represents approximately 5-10% of all patients with breast cancer. This syndrome is mainly associated to high-risk pathogenic alleles in BRCA1 and BRCA2 genes, but only for 25% of HBOC cases. This work aimed to find new pathogenic alleles in a panel of 143 cancer-predisposing genes in 300 Mexican cancer patients with suspicion of HBOC and 31 non-cancer patients with a severe family history of cancer, using massive parallel sequencing. We found 15% (45/300) patients with pathogenic variants in the group of cancer patients; 12% (35/300) harbored variants with unknown clinical significance (VUS) and 73% (220/300) were negative. The non-cancer group had a 32% (10/31) of patients with pathogenic variants, 3% (1/31) had VUS and 65% (20/31) were negative. Moreover, the most recurrent mutation was the Mexican founder deletion of exons 9-12 in BRCA1, found in 5 of 16 cancer patients with alterations in this gene. Private or rare VUS variants with potential impact at protein level were found in 22 genes, being CHEK2 the one with most VUS (6/39). Noteworthy, our results show for the first time in the Mexican population an equal contribution of pathogenic alleles in other susceptibility cancer genes (50%) as in BRCA1/2 (50%). This highlights the high locus heterogeneity of HBOC and the necessity of expanding genetic tests for this disease to broader gene panels. Further studies need to be conducted to define the clinical impact of the pathogenic alleles and VUS identified. Citation Format: Felipe Vaca-Paniagua, Rosalía Quezada-Urban, Clara E. Díaz-Velásquez, Rina Gitler, María P. Rojo-Castillo, Max Sirota-Toporek, Andrea Figueroa-Morales, Oscar Moreno-García, Lizbeth García Esquivel, Gabriela Torres-Mejía, Michael Dean, Ivan Delgado-Enciso, Héctor Ochoa-Díaz-López, Fernando Rodriguez-León, Virginia Jan, Victor H. Hugo Garzón-Barrientos, Pablo Ruiz-Flores, Perla K. Espino-Silva, Jorge Haro-Santa Cruz, Héctor Martínez-Gregorio, Ernesto Rojas-Jiménez, Rosa M. Álvarez-Gómez, Luis A. Herrera, Isabelle Romieu, Luis I. Terrazas, Yolanda I. Chirino, Cecilia Frecha, Javier Oliver, Sandra Perdomo. Comprehensive analysis of germline variants in Mexican patients with hereditary breast and ovarian cancer susceptibility [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1240.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 9 ( 2017-05-01), p. 2266-2278
    Abstract: In glioblastoma (GBM), tumor-associated macrophages (TAM) represent up to one half of the cells of the tumor mass, including both infiltrating macrophages and resident brain microglia. In an effort to delineate the temporal and spatial dynamics of TAM composition during gliomagenesis, we used genetically engineered and GL261-induced mouse models in combination with CX3CR1GFP/WT;CCR2RFP/WT double knock-in mice. Using this approach, we demonstrated that CX3CR1LoCCR2Hi monocytes were recruited to the GBM, where they transitioned to CX3CR1HiCCR2Lo macrophages and CX3CR1HiCCR2− microglia-like cells. Infiltrating macrophages/monocytes constituted approximately 85% of the total TAM population, with resident microglia accounting for the approximately 15% remaining. Bone marrow–derived infiltrating macrophages/monocytes were recruited to the tumor early during GBM initiation, where they localized preferentially to perivascular areas. In contrast, resident microglia were localized mainly to peritumoral regions. RNA-sequencing analyses revealed differential gene expression patterns unique to infiltrating and resident cells, suggesting unique functions for each TAM population. Notably, limiting monocyte infiltration via genetic Ccl2 reduction prolonged the survival of tumor-bearing mice. Our findings illuminate the unique composition and functions of infiltrating and resident myeloid cells in GBM, establishing a rationale to target infiltrating cells in this neoplasm. Cancer Res; 77(9); 2266–78. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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...