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)  (11)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 4_Supplement ( 2022-02-15), p. P3-07-02-P3-07-02
    Abstract: Background Detection of pathogenic GVs in patients with BC has implications for both patients and their family members. Management options such as increased surveillance, chemoprevention, and surgical prophylaxis are available to GV carriers. Beyond BRCA1/2, GVs in the cancer susceptibility genes (CSGs) PALB2, ATM, and CHEK2 confer a 2-11-fold lifetime risk of BC. One opportunity CGP assays present is the potential to detect clinically-relevant GVs in addition to targetable somatic variants. While the Breast Cancer Association Consortium found that 5.2% of women with BC carried a GV in one of these five CSGs, we sought to describe the frequency of these potential GVs detected by CGP in a cohort of patients with advanced disease. Methods We reviewed an internal database of patients with advanced BC who underwent testing with a CGP panel using tissue (n= 20,109, FoundationOne® CDx) or plasma (n= 4,182, FoundationOne®Liquid CDx or FoundationOne®Liquid). Cases with a potential GV were identified by filtering base substitutions and short indels for inclusion in ClinVar as pathogenic or likely pathogenic and by variant allele frequency (VAF) based on an optimized assay-specific threshold, focusing on the select CSGs of BRCA1/2, PALB2, ATM and CHEK2. To enable follow-up of potential GVs, we implemented a new reporting “banner” to highlight select short variants in these CSGs. Predominant patient ancestry was inferred using a SNP-based classifier and Fisher’s exact test was utilized for comparison between groups. Results A total of 24,291 unique patients with primarily advanced BC had CGP results available for study, with common actionable findings including PIK3CA mutations (8,572, 35.3%), ESR1 mutations (3,289, 13.5%), and HER2 amplification (1,602, 6.6%). Focusing on the 5 CSGs, 16.4% of patients (3,986) had at least one pathogenic alteration detected in BRCA2 (1,153, 4.7%), ATM (969, 4.0%), CHEK2 (982, 4.0%) BRCA1 (849, 3.5%) or PALB2 (308, 1.3%); 1.1% of patients (263) harbored alterations in multiple CSGs. 50.7% (2,020/3,986) of patients with pathogenic alterations in these CSGs - 8.3% (2,020/24,291) of total patients with advanced BC - had an alteration meeting criterion as a potential GV. Variants in BRCA1 (511/919, 55.6%), BRCA2 (786/1,425, 55.2%), and PALB2 (201/376, 53.5%), more frequently met criteria as potential GVs than variants in CHEK2 (328/1,046, 31.4%) or ATM (265/1,100, 24.1%). In these five CSGs, 1,796/3,195 alterations detected on tissue CGP (56.2%) and 295/1,671 detected in plasma (17.7%) met criteria as a potential GVs. Ancestry analysis of 20,108 assessable BC patients tested using tissue CGP showed potential GVs in CHEK2 were more common in European vs non-European (1.7% vs 0.4%, p & lt;0.01) and potential GVs in PALB2 were more common in African vs non-African (1.4% vs 0.7%, p & lt;0.01) ancestries. Of 1,961 patients with BC tested over a 2-month period, 9.7% of reports (191) included a germline banner reporting a potential GV in one of these 5 CSGs and recommending consideration of referral for germline testing. Conclusion Potential pathogenic GVs in BRCA1/2, PALB2, ATM and CHEK2 were identified in 8.3% of patients with advanced BC tested utilizing CGP when filtering by VAF and ClinVar annotation. Highlighting these potential GVs with a report banner provides the opportunity for follow-up germline testing and genetic counseling for patients who otherwise may not have been referred for additional testing. These alterations were detected in both tissue and plasma CGP and in patients of varying ancestries. The potential for detection of potential GVs plus the detection of actionable driver and resistance mechanisms may add to the clinical value of CGP for patients with breast cancer. Citation Format: Marni B Tierno, Kali C Dougherty, Erica Gornstein, Dean C Pavlick, Alexa Schrock, Geoff R Oxnard. Identification of potential germline variants (GV) on tumor comprehensive genomic profiling (CGP) in patients with advanced breast cancer (BC): BRCA1/2 and beyond [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 P3-07-02.
    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 ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 4_Supplement ( 2022-02-15), p. PD9-10-PD9-10
    Abstract: Purpose: Patients with triple negative breast cancer (TNBC) with residual disease after neoadjuvant chemotherapy (NAC) have high risk of recurrence with prior data suggesting improved outcomes with capecitabine. Targeted agents have demonstrated activity across multiple cancer types. BRE12-158 was a phase 2, multicenter trial that randomized TNBC patients with residual disease after NAC to genomically-directed therapy vs. treatment of physician choice (TPC). Patients and Methods: From March 2014 to December 2018, 197 patients were enrolled. Residual tumors were sequenced using a next generation sequencing (NGS) test. A molecular tumor board adjudicated all results. Patients were randomized to 4 cycles of genomically-directed therapy (arm A) vs. TPC (arm B). Patients without a target were assigned to arm B. Primary endpoint was 2-year disease free survival (DFS) among randomized patients. Secondary/exploratory endpoints included: distant disease free survival (DDFS), overall survival (OS), toxicity assessment, time-based evolution of therapy, and drug-specific outcomes. Results: 193 patients were randomized or were assigned to arm B. The estimated 2-year DFS was 56.6% (95%CI:0.45-0.70) for arm A vs. 62.4% (95%CI:0.52-0.75) for randomized arm B. No difference was seen in DFS, DDFS, or OS for the entire or randomized populations. There was increased uptake of capecitabine for TPC over time. Patients randomized later had less distant recurrences. ctDNA status remained a significant predictor of outcome with some patients demonstrating clearance with post-neoadjuvant therapy. Conclusion: Genomically directed therapy was not superior to TPC for patients with residual TNBC after NAC. Capecitabine should remain the standard of care; however, the activity of other agents in this setting provides rationale for testing optimal combinations to improve outcomes for this high-risk population. ctDNA should be considered a standard covariate for trials in this setting. Citation Format: Bryan P Schneider, Guanglong Jiang, Tarah J Ballinger, Fei Shen, Christopher Chitambar, Rita Nanda, Carla Falkson, Filipa C Lynce, Christopher Gallagher, Claudine Isaacs, Marcelo Blaya, Elisavet Paplomata, Radhika Walling, Karen Daily, Reshma Mahtani, Michael A Thompson, Robert Graham, Maureen E Cooper, Dean C Pavlick, Lee A Albacker, Jeffery Gregg, Jeffery P Solzak, Yu-Hsiang Chen, Casey L Bales, Erica Cantor, Bradley A Hancock, Nawal Kassem, Paul Helft, Bert O'Neil, Anna Maria Storniolo, Sunil Badve, Kathy D Miller, Milan Radovich. BRE12-158: A post-neoadjuvant, randomized phase 2 trial of personalized therapy vs. treatment of physician’s choice for patients with residual triple negative breast cancer [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 PD9-10.
    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 ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 4_Supplement ( 2020-02-15), p. GS5-02-GS5-02
    Abstract: Background: A significant proportion of patients with early-stage TNBC are treated with neoadjuvant chemotherapy (NAC). Sequencing of ctDNA after surgery can be used to detect minimal residual disease and predict which patients may experience clinical recurrence. Methods: BRE12-158 is a recently completed Phase II clinical trial which randomized early-stage TNBC patients with residual disease after NAC to post-neoadjuvant genomically-directed therapy vs treatment of physician choice. 151 patients had a plasma sample collected at the time of treatment assignment (after surgery and radiation). ctDNA was successfully sequenced in 150 patients. 148 of the 150 sequenced patients had clinical follow-up. Sequencing was performed by Foundation Medicine using the FoundationOne Liquid assay which profiles for 70 commonly mutated oncogenes. Presence of mutated ctDNA was associated with distant disease free survival (DDFS) and overall survival (OS) in univariate analysis using the Log-Rank test, and in multi-variate analysis using Cox proportional hazards model. Results: Mutated ctDNA was detected in 94 of 148 sequenced patients (64%). TP53 was the most commonly mutated gene consistent with prior genomic studies of TNBC. At 16.7 months of median follow-up, detection of ctDNA was significantly associated with an inferior DDFS (median DDFS 32.5 months vs. Not Reached, p=0.0030). At 24 months, the DDFS probability was 53% in ctDNA-positive patients as compared to 81% in ctDNA-negative patients. In multi-variate analysis, when considering significant covariates, including: residual cancer burden (RCB); number of positive lymph nodes; tumor size; stage; grade; age; and race; detection of ctDNA remained independently associated with inferior DDFS (HR=3.1, CI: 1.4-6.8, p=0.0048). Similarly, detection of ctDNA was associated with inferior OS in univariate (p=0.021) and multi-variate analysis (HR=2.7, CI:1.1-6.2, p=0.022). Lastly, we observed a correlation between higher maximum somatic allele frequency and a shorter DDFS interval in multivariate analysis (HR=4.7, CI: 1.04-21.1, p=0.044) and shorter OS (HR=4.9, CI:1.06-22.4, p=0.041), suggesting that the quantitative degree of ctDNA burden is associated with clinical outcome. Conclusions: Detection of ctDNA in early-stage TNBC after neoadjuvant chemotherapy is an independent predictor of disease recurrence, and represents an important novel stratification factor for future post-neoadjuvant trials. Citation Format: Milan Radovich, Guanglong Jiang, Christopher Chitambar, Rita Nanda, Carla Falkson, Filipa C. Lynce, Christopher Gallagher, Claudine Isaacs, Marcelo Blaya, Elisavet Paplomata, Radhika Walling, Karen Daily, Reshma Mahtani, Michael A. Thompson, Robert Graham, Maureen E. Cooper, Dean C. Pavlick, Lee Albacker, Jeff Gregg, Casey L. Bales, Bradley A. Hancock, Erica Cantor, Fei Shen, Anna Maria V. Storniolo, Sunil Badve, Tarah Ballinger, Kathy D. Miller, Bryan P. Schneider. Detection of circulating tumor DNA (ctDNA) after neoadjuvant chemotherapy is significantly associated with disease recurrence in early-stage triple-negative breast cancer (TNBC): Preplanned correlative results from clinical trial BRE12-158 [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 GS5-02.
    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 ...
  • 4
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 14, No. 12_Supplement_2 ( 2015-12-01), p. PR05-PR05
    Abstract: Genomic assessment of exceptional responders is a promising approach to identify predictors of response to antibody therapy directed against the immune checkpoint programmed death 1 (PD-1) receptor, which has been shown to yield prolonged and deep responses in multiple types of human cancer. We identified a patient with endometrial cancer who experienced an exceptional response to pembrolizumab, an antibody to programmed death 1 (PD-1) receptor. The primary endometrial cancer specimen and the biopsy from the recurrent supraclavicular lymph node (LN) metastasis obtained prior to treatment were analyzed by hybrid-capture based genomic profiling at a commercial CLIA-certified laboratory, Foundation Medicine, targeting all exons of 315 cancer-related genes. In the patient's pre-treatment endometrial cancer specimens we identified a mutation in DNA polymerase epsilon gene (POLE), which is associated with disruption of the exonuclease activity required for proofreading function and results in a high mutation burden or “ultramutator” phenotype. This tumor did harbor a large number of mutations: 32 likely pathogenic sequence variants and 116 variants of unknown significance (VUS). We next reviewed genomic alterations in 252 deidentified endometrioid endometrial cancers that underwent genomic profiling with the FoundationOne assay and determined that 23 (9.1%) had sequence variants in POLE. The cancers with POLE sequence variants had a mean of 21.2 +/-4.1 mutations identified as likely pathogenic and 82.2 +/-25 variants identified as VUS, compared with a mean of 7.5+/-0.5 likely pathogenic variants and 12.8 +/- 2.6 VUS in POLE wt cases (mean +/- S.E.; p & lt;0.005 and P = 0.015, respectively). This is consistent with TCGA data showing that POLE mutant cancers typically harbor an extremely high mutational burden. To determine if POLE mutant cancers were associated with an immune signature, analysis of RNA sequencing data from endometrioid endometrial cancers in TCGA was performed. POLE mutant cancers have higher expression of several genes encoding for immune checkpoint-related proteins, including PD-L1 and PD-L2, than either MSI or MSS endometrioid cancers. POLE mutant cancers also showed higher expression of T-cell markers such as CD8A, CD3G, PD-1 and CTLA-4, suggesting the presence of a pre-existing T-cell infiltrate. Analysis of histologic image data from TCGA confirmed that POLE mutant cancers had presence of a robust lymphocytic infiltrate. These data suggest that endometrial cancers harboring POLE mutations are associated with expression of immune checkpoint genes and evidence of lymphocytic infiltration. Thus, these tumors may be exceptionally vulnerable to treatment with immune checkpoint inhibitor therapy. We propose further clinical investigation with immunotherapy in endometrial and other cancers with POLE mutations. Citation Format: Janice M. Mehnert, Anshuman Panda, Hua Zhong, Kim M. Hirshfield, Sherri Damare, Katherine Stiles, Levi Sokol, Mark N. Stein, Lorna Rodriguez-Rodriguez, Howard L. Kaufman, Siraj Ali, Jeffery Ross, Dean C. Pavlick, Gyan Bhanot, Eileen P. White, Robert S. DiPaola, Ann Lovell, Jonathan Cheng, Shridar Ganesan. Exceptional Response to PD-1 antibody treatment in a POLE-mutant endometrial cancer. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr PR05.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 4_Supplement ( 2021-02-15), p. PS5-04-PS5-04
    Abstract: Background: Non-colorectal MSI-H tumors are increasingly identified by CGP. Rare types such as MSI-H BC remain poorly defined with an evidence gap on how to optimally sequence or combine with standard of care treatment. MSI can be measured by either IHC, PCR, or CGP and can be caused by both sporadic and germline variants within different tumor types. Prior studies in BC have shown evidence of dMMR by IHC cases MSS based on PCR. This could be due to intra-tumor heterogeneity, specific microsatellite loci evaluated, or penetrance of germline, somatic, or epigenetic alterations. Published data suggests carriers of germline pathogenic MMR variants have a BC risk equivalent to the normal population and currently germline testing is recommended only for BRCA. Currently in advanced BC, standard tumor biomarker testing includes IHC, PCR, and FISH; however, with increasing use of CGP we demonstrate additional actionable biomarkers as well as potential germline variants in MSI-H BC. Methods: DNA was extracted and hybrid capture CGP was performed on 29,160 BC cases. TMB was determined on 0.8-1.2 Mb of DNA and MSI status on 95-114 loci. Genomic LOH was also evaluated. Comparative analysis was done with 101 MSI-H BC, 841 MSS BC and 4,988 non-breast MSI-H cancers. Histological subtype was obtained from the pathology along with orthogonal testing for ER/PR/HER2 status. Somatic-germline-zygosity (SGZ) status was predicted using a published research use algorithm. Select case reports with clinical outcomes will be presented. Results: We identified 101 (0.35% of total) MSI-H BC cases: 29 ER+/HER2-, 5 HER2+, 29 TNBC, and 28 unknown. Amongst BC cases with known subtype, TNBC was enriched for MSI-H vs MSS (53.4 vs 35.8%, p=0.005). The median TMB in MSI-H BC (26.1 mut/Mb, IQR 17.4;42.8) was significantly lower than that of MSI-H colon (46.1mut/MB) and higher than that of MSI-H uterine tumors (22.6mut/Mb) in our comparison group (p & lt;0.001 for both, Kruskal-Wallis test). Pathogenic variants in an MMR gene were found in 61.4% of MSI-H BC with MLH1 loss being the most common (13.6%) and much higher vs. the non-breast MSI-H cohort (2.4%, p & lt;0.0001). Germline mutations in MMR genes in BC are rare yet 5/52 MMR short variants identified in 101 MSI-H BCs were predicted to be germline, 34 somatic, and 13 could not be determined. We identified 21 MSI-H BC patients with a total of 25 pathogenic BRCA1/2 alterations of which 4 were likely germline, 10 were homozygous, and were enriched in TNBC. These were mainly frameshift mutations, including BRCA2 T3033fs* in 5/18 (28%) cases; however, 7/25 were deletions, rearrangements, or nonsense mutations. Median gLOH was significantly higher in BRCA altered (19.7%) compared to BRCA wild-type MSI-H BC cases (9.6%) (p=0.007, Wilcox test). Additional potentially targetable biomarkers included 26 CDx eligible PIK3CA mutations, 11 ERBB2 activating point mutations in the TKD or ECD domain, 1 FGFR2 rearrangement, and 6 AKT1 E17K mutations. Four cases also had concurrent (CD274) PD-L1 amplifications. Conclusion: MSI-H BC is rare but CGP can identify additional therapeutic options for rational combination with targeted therapies such as PI3K, PARP, and HER2 inhibitors. BRCA alterations may be of germline or somatic origin and they may be targetable, as demonstrated by gLOH, rather than passenger mutations. Further characterization of these tumors and comparison to both MSS BC and non-breast MSI-H tumor types, combined with treatment outcomes, can provide insights on rationale combinations and/or sequencing of therapeutic agents. Citation Format: Kimberly McGregor, Natalie Danzinger, Jeffrey S. Ross, Kyle Gowen, Alexa B. Schrock, Garrett M. Frampton, Dean C. Pavlick, Jan W. Davis, Carl R. Gray, Jeffrey M. Venstrom. Therapeutic considerations in microsatellite instability high (MSI- H) breast cancers (BC) identified by comprehensive genomic profiling (CGP) [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS5-04.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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. 82, No. 12_Supplement ( 2022-06-15), p. 2139-2139
    Abstract: Tumor mutational burden (TMB) derived from tissue biopsies (tTMB) has been associated with clinical efficacy in patients treated with immune checkpoint inhibitors; the clinical utility of TMB derived from blood samples (bTMB) has also been demonstrated but is less widely studied. Little is known about the concordance between tissue- and blood-derived genomic assessments and the factors that contribute to their discordance, underlying the need for further investigation of sequence alteration profiles for successful adoption of noninvasive tumor profiling. We explored the genomic landscape, including concordance between tTMB and bTMB, in samples from patients who were screened for enrollment into CheckMate 848, a prospective, phase 2 study of nivolumab plus ipilimumab and nivolumab monotherapy in patients with advanced or metastatic solid tumors with high TMB (NCT03668119). Of 1954 screened patients, 212 were randomized, with a cap of 15% per tumor type. Tissue- (FoundationOne® CDx—based Clinical Trial Assay) and blood-based (Foundation Medicine bTMB Clinical Trial Assay) genomic data were utilized for analysis of genomic variants, tTMB, and bTMB. In total, 1438 unique tissue and 1720 unique plasma samples were analyzed during trial screening (June 2021 database lock). Of over 100 screened disease ontologies (taken from the tissue diagnosis), pancreatic (9.7%), breast (8.8%), and ovarian (6.1%) cancers, as well as cholangiocarcinoma (5.2%), were the most common. A total of 1141 tissue and 1573 plasma samples passed established quality control criteria, resulting in ascertainment levels of 79.3% for tTMB and 91.5% for bTMB. A correlation between tTMB and bTMB scores was identified across 1017 tissue and plasma sample pairs (Spearman’s r, 0.48; P & lt; 0.0001). Median (range) tTMB and bTMB were 3.8 (0—452.6) and 3.5 (0—1027.5) mutations per megabase (mut/Mb), respectively. High microsatellite instability (MSI) was detected in 25 (2.5%) MSI-evaluable tissue samples; in these patients, median tTMB was 25.2 mut/Mb. At the prespecified cutoff of 10 mut/Mb, 15.8% and 20.7% of samples had high tTMB and bTMB, respectively; the positive (PPA), negative, and overall percentage agreements between assays were 60%, 88%, and 84%, respectively. TMB correlation (Spearman’s r, 0.54; P & lt; 0.0001) and PPA (66%) were improved among 806 (79.3%) sample pairs with plasma maximum somatic allele frequency ≥ 1%. In CheckMate 848, data from paired biopsies revealed the complementary nature of TMB assessments from tissue and blood, suggesting that both approaches may have the potential to identify high mutational burden in samples obtained from patients with advanced solid tumors. Further interrogation of the biological and analytical factors affecting tumor- and blood-derived genomic profiling is warranted to support their implementation in clinical settings. Citation Format: Jie He, Natallia Kalinava, Parul Doshi, Jie Ma, Dean C. Pavlick, Lee A. Albacker, Hanna Tukachinsky, Gina Fusaro, Geoffrey R. Oxnard, George Green, David Fabrizio, Jonathan Baden. Evaluation of tissue- and plasma-derived tumor mutational burden and genomic alterations of interest from the CheckMate 848 clinical trial [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 2139.
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 4_Supplement ( 2022-02-15), p. P5-08-16-P5-08-16
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 4_Supplement ( 2022-02-15), p. P5-08-16-P5-08-16
    Abstract: Background: Homozygous deletion of the MTAP gene (MTAPdel) results in arginine accumulation in tumor cells and is currently being used for patient selection in clinical trials testing novel inhibitors in several tumor types. Methods: 7,308 clinically advanced ductal and lobular breast cancers (MBC) underwent hybrid-capture based comprehensive genomic profiling to evaluate all classes of genomic alterations (GA). Tumor mutational burden (TMB) was determined on up to 1.1 Mbp of sequenced DNA and microsatellite instability (MSI) was determined on up to 114 loci. PD-L1 tumor cell expression was determined by IHC (Ventana SP142 immunocyte scoring assay used in 81% or Dako 22C3 tumor cell scoring assay used in 19%). ER and PR staining as reported by treating facility was available for a subset of 1,204 cases. HER2 status for these cases was assigned based on the presence of ERBB2 amplification (amp). All cases were of known lobular (ILC) or ductal (IDC) subtype. Results: 212 (2.9%) MBC featured MTAPdel. When compared to MTAPintact MBC, MTAPdel MBC were younger (p=.004) and less often of lobular subtype (p & lt;.0001). They were less frequently ER and PR positive and more frequently triple negative (TNBC) (both p & lt;.0001). ERBB2 amp frequencies were similar. The GA/tumor were similar when CDKN2A/B del (near universally present on MTAPdel cases) are excluded from analysis. Of non-targetable GA, TP53 GA were more frequent in MTAPdel MBC. In the MTAPintact group, ESR1 GA were more common, likely reflecting use of hormonal therapy given the greater frequency of ER/PR+ cases, and RB1 GA were similarly more frequent possibly reflecting use of cell cycle inhibitors. CDH1 GA reflected the greater ILC frequency in the MTAPintact group. For the currently targetable GA, the MTAPdel MBC featured more frequent BRCA1 and PTEN GA and the MTAPintact MBC had more ERBB2 non-amplification GA, PIK3CA and CCND1 GA. The immunotherapy predictive biomarkers microsatellite instability (MSI) and tumor mutational burden (TMB) were similarly low in both groups, but CD274 (PD-L1) amplification and PD-L1 expression were higher in the MTAPdel patients. Conclusions: MTAPdel MBC differs significantly from MTAPintact MBC in routine clinical features and GA impacting both targeted and immunotherapies. Given the current early and mid-stage development of MTA2 and PRMT5 inhibitors that exploit a synthetic lethality pathway for MTAPdel cancer, further study of selecting MTAPdel MBC for clinical trials appears warranted. MTAPIntact MBCMTAPdel MBC P valueNumber of Cases7096212Age (median + range)59 (22-89+)54 (25-89)Age (mean)58.155.6=.004ILC Prevalence15.2%1.4% & lt;.0001ER+69.2%*49%^ & lt;.0001PR+49.1%*34%^ & lt;.0001HER2+ (ERBB2 amplification +)7.8%7.4%NSTNBC27.1%*45.3%^ & lt;.0001GA/tumor with CDKN2A/B excluded6.16.6NSUntargetable GACDKN2A3.1%100% & lt;.0001CDKN2B1.3%97.4% & lt;.0001TP5352.1%62.4%=.004ESR18.5%3.7%=.011RB17.3%1.1% & lt;.0001CDH114.5%1.1% & lt;.0001Potentially Targetable GAERBB2 non-amplification3.0%0%=.003PIK3CA36.6%23.3%=.001CCND117.8%0.5% & lt;.0001BRCA13.7%10.1% & lt;.0001BRCA24.2%5.8%NSFGFR113.4%11.1%NSPTEN13.2%22.8%=.0002NF16.4%8.5%NSImmunotherapy BiomarkersCD274 amplification1.4%3.2%=.03MSI-H & lt;1% & lt;1%NSMedian TMB2.52.5NSTMB ≥ 10 mut/Mb7.9%5.1%NSPD-L1 Low3.9% (of 2895 cases)13% (of 92 cases)=.0004PD-L1 High0.7% (of 2895 cases)0% (of 92 cases)NS*Based on 1,014 cases ^Based on 190 cases Citation Format: Lajos Pusztai, Natalie Danziger, Ethan S Sokol, Dean C Pavlick, Ryon Graf, Shakti Ramkissoon, Richard SP Huang, Brennan J Decker, Jeffrey S Ross. MTAP deletion and synthetic lethality-based drug development for metastatic breast cancer (MBC) [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 P5-08-16.
    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 ...
  • 8
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 12_Supplement ( 2019-12-01), p. B091-B091
    Abstract: Introduction: One major form of vulvar squamous cell carcinoma (vSCC) is associated with detectable high-risk strains of human papillomavirus (hrHPV) and is often accompanied by usual-type vulvar intraepithelial neoplasia (VIN). The second major form of vSCC is often associated with chronic dystrophic or inflammatory lesions in postmenopausal women, does not harbor detectable HPV infection, and is often preceded by p53-mutant differentiated VIN. While studies have examined the two subtypes, no large-scale genomic study has been performed to our knowledge. We sought to assess the genomics of a large cohort of aggressive vSCCs, with an aim to identify distinct mutational signatures based on the presence or absence of hrHPV genome reads. Methods: 280 vSCC were tested by hybridization capture of up to 406 cancer-related genes evaluated for base substitutions, small indels, amplification (amp), and rearrangements. HPV genome sequences were detected by de novo assembly of non-human sequencing reads and BLASTn comparison against all viral nucleotide sequences in the NCBI RefSeq database. Tumor mutational burden (TMB, mutations/Mb) was determined on ~1.1 Mbp of sequenced DNA. PD-L1 status was determined by IHC (Dako 22C3), with ≥50% tumor proportion score defined as high positive. Results: 102/280 vSCCs contained hrHPV sequences. Of these, 90 were HPV-16, 7 HPV-18, 1 HPV-31, 3 HPV-33, 1 HPV-58, and 1 HPV-67. Patients were significantly younger in the HPV(+) group (median 59 v. 64 years, p=0.001). Compared with the HPV(–) cohort, HPV(+) cases showed significantly more pathogenic genomic alterations (GA) in PIK3CA (31% vs. 17%, p=0.004), PTEN (14% vs. 2%, p & lt;0.0001), EP300 (14% vs. 1%, p & lt;0.0001), STK11 (14% vs. 1%, p & lt;0.0001), AR (5% vs. 0%, p=0.006), and FBXW7 (10% vs. 3%, p=0.03). In contrast, HPV(–) cases showed significantly more alterations in TP53 (82% vs. 3%, p & lt;0.0001), TERTp (71% vs. 8%, p & lt;0.0001), CDKN2A (55% vs. 2%, p & lt;0.0001), CCND1 (23% vs. 2%, p & lt;0.0001), FAT1 (25% vs. 4%, p & lt;0.0001), NOTCH1 (19% vs. 6%, p=0.002), and EGFR (amp: 12% vs. 0%, p & lt;0.0001), as well as a higher rate of 9p24.1 (PDL1/PDL2) amp (7% vs. 1%) and PD-L1 IHC high-positive tumor staining (33% vs. 9%, p=0.04). Differences in alterations were observed between known primary and metastatic sites in cases with similar HPV status but did not reach significance (table). HPV(+)HPV(–) PrimaryMetastasisPrimaryMetastasis # of cases504112442 Age (range)58 (36-81)60 (29-83)64 (25-89)63 (45-89) Median TMB (range)5.2 (0-18.3)6.1 (0-47.8)3.5 (0-90.5)5.0 (0-13) PIK3CA GA26%37%17%17% PTEN GA18%10%2%2% STK11 GA10%22%1%2% FBXW7 GA10%10%5%0% TP53 GA0%5%82%81% TERTp GA6%10%73%64% CDKN2A GA0%5%55%52% CCND1 GA2%2%20%29% EGFR amp0%0%12%14% CD274(PD-L1) amp*0%0%6%10% PD-L1 IHC high*8%11%30%33% TMB & gt;10*6%24%3%19% Conclusions: vSCCs show significant differences in molecular profile based on HPV status. 63% of metastatic HPV(+) cases (54% overall) have a potentially actionable alteration in the PI3K/mTOR pathway, and 42% of metastatic HPV(–) cases (39% overall) have at least one potential predictive biomarker* for response to immunotherapy. Our findings provide compelling rationale for tandem comprehensive genomic profiling and HPV assessment of advanced vulvar SCCs to more fully inform therapeutic options and stratification in clinical trials. Citation Format: Erik A Williams, Adrienne J Werth, Meagan Montesion, Ethan S Sokol, Dean C Pavlick, Nikunj A Shah, Jo-Anne Vergilio, Natalie A Danziger, Jonathan K Killian, Douglas A Lin, Vincent A Miller, Jeffrey S Ross, Julia A Elvin. Vulvar squamous cell carcinoma: Comprehensive genomic profiling of HPV(+) versus HPV(–) forms reveals a different set of potentially actionable biomarkers [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr B091. doi:10.1158/1535-7163.TARG-19-B091
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 4_Supplement ( 2020-02-15), p. P4-06-07-P4-06-07
    Abstract: Introduction: De-regulation of the class I phosphoinositide 3’-kinase (PI3K) pathway has long been known to contribute to the development and progression of many tumors. However, the FDA only recently approved the first selective inhibitor of PIK3CA, the p110-alpha catalytic subunit of PI3K, specifically for use in treatment of a subset of PIK3CA-mutant breast carcinomas. Given the emergence of this therapeutic class of agents, we sought to identify other subsets of breast tumors that may be driven largely by PIK3CA mutations and which therefore could be candidates for these therapies. In breast fibroepithelial neoplasms, mutations in PIK3CA have been occasionally reported in borderline and malignant phyllodes tumors. In contrast, mutations in MED12 are common and recurrent across the entire spectrum of benign and malignant breast fibroepithelial tumors, and are enriched in borderline/malignant phyllodes cases that still have benign fibroadenoma-like areas. In the current study, we sought to define the histologic and molecular features of PIK3CA-mutant phyllodes tumors. Methods: From 2014 to 2019, we analyzed clinical tumor samples using comprehensive genomic profiling by a hybrid capture-based DNA sequencing platform. We searched our case archive to find breast phyllodes tumors with known or likely pathogenic alterations in PIK3CA and other known tumor-related genes. All cases were clinically advanced. We reviewed pathology reports, histopathology, and patient clinical data. Results: We identified 12 (16%) of 76 breast phyllodes tumors in our case archive as PIK3CA-mutant. Median patient age for PIK3CA-mutant phyllodes tumors was 56 years. Cases consisted of 6 primary tumors, 2 local recurrences, and 4 lung metastases. Primary tumor size measured from 38 to 220 mm (median 100 mm; mean 114 mm). Digital slides were available for histology review in 9 cases. Cases showed uniformly malignant histology, with no benign or fibroadenoma-like regions. 3 cases showed malignant heterologous elements. Compared to the rest of our breast phyllodes tumor cohort, PIK3CA-mutant cases showed significantly fewer pathogenic genomic alterations in MED12 (8% vs. 55%, p=0.0037) and TP53 (17% vs. 56%, p=0.0245), as well as a trend to fewer mutations in RB1 (0% vs. 22%, p=0.11). The other most frequently mutated genes in the PIK3CA-mutant group were TERTp (70%), CDKN2A (67%), and NF1 (50%). Conclusions: PIK3CA-mutant phyllodes tumors define a unique subset of tumors characterized by aggressive histologic features and largely lacking the MED12 mutations seen in many fibroepithelial neoplasms. These findings provide compelling rationale for comprehensive genomic profiling of advanced cases of this disease in an effort to inform therapeutic options including clinical trials of PI3K-targeted agents in this setting. Citation Format: Erik A Williams, Ethan S Sokol, Dean C Pavlick, Nikunj Shah, Julia A Elvin, Jo-Anne Vergilio, Jonathan K Killian, Nhu Ngo, Douglas Lin, Vincent A Miller, Jeffrey S Ross. PIK3CA-mutant breast phyllodes tumors show a uniformly aggressive histology and significant mutual exclusivity with MED12 mutation [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 P4-06-07.
    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 ...
  • 10
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 11, No. 2 ( 2021-02-01), p. 282-292
    Abstract: Neoantigen presentation arises as a result of tumor-specific mutations and is a critical component of immune surveillance that can be abrogated by somatic LOH of the human leukocyte antigen class I (HLA-I) locus. To understand the role of HLA-I LOH in oncogenesis and treatment, we utilized a pan-cancer genomic dataset of 83,644 patient samples, a small subset of which had treatment outcomes with immune checkpoint inhibitors (ICI). HLA-I LOH was common (17%) and unexpectedly had a nonlinear relationship with tumor mutational burden (TMB). HLA-I LOH was frequent at intermediate TMB, yet prevalence decreased above 30 mutations/megabase, suggesting highly mutated tumors require alternate immune evasion mechanisms. In ICI-treated patients with nonsquamous non–small cell lung cancer, HLA-I LOH was a significant negative predictor of overall survival. Survival prediction improved when combined with TMB, suggesting TMB with HLA-I LOH may better identify patients likely to benefit from ICIs. Significance: This work shows the pan-cancer landscape of HLA-I LOH, revealing an unexpected “Goldilocks” relationship between HLA-I LOH and TMB, and demonstrates HLA-I LOH as a significant negative predictor of outcomes after ICI treatment. These data informed a combined predictor of outcomes after ICI and have implications for tumor vaccine development. This article is highlighted in the In This Issue feature, p. 211
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...