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)  (10)
  • Park, Se Hoon  (10)
Material
Publisher
  • American Association for Cancer Research (AACR)  (10)
Language
Years
Subjects(RVK)
  • 1
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 9, No. 10 ( 2019-10-01), p. 1388-1405
    Abstract: The VIKTORY (targeted agent eValuation In gastric cancer basket KORea) trial was designed to classify patients with metastatic gastric cancer based on clinical sequencing and focused on eight different biomarker groups (RAS aberration, TP53 mutation, PIK3CA mutation/amplification, MET amplification, MET overexpression, all negative, TSC2 deficient, or RICTOR amplification) to assign patients to one of the 10 associated clinical trials in second-line (2L) treatment. Capivasertib (AKT inhibitor), savolitinib (MET inhibitor), selumetinib (MEK inhibitor), adavosertib (WEE1 inhibitor), and vistusertib (TORC inhibitor) were tested with or without chemotherapy. Seven hundred seventy-two patients with gastric cancer were enrolled, and sequencing was successfully achieved in 715 patients (92.6%). When molecular screening was linked to seamless immediate access to parallel matched trials, 14.7% of patients received biomarker-assigned drug treatment. The biomarker-assigned treatment cohort had encouraging response rates and survival when compared with conventional 2L chemotherapy. Circulating tumor (ctDNA) analysis demonstrated good correlation between high MET copy number by ctDNA and response to savolitinib. Significance: Prospective clinical sequencing revealed that baseline heterogeneity between tumor samples from different patients affected response to biomarker-selected therapies. VIKTORY is the first and largest platform study in gastric cancer and supports both the feasibility of tumor profiling and its clinical utility. This article is highlighted in the In This Issue feature, p. 1325
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2607892-2
    detail.hit.zdb_id: 2625242-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 2912-2912
    Abstract: Recently, the sidedness of the primary tumor (right versus left) has been investigated for its ability to prognosticate and predict outcomes. We evaluated the effect of regorafenib based on KRAS mutation status and the sidedness of the primary tumor in patients with metastatic colorectal cancer (mCRC). We analyzed 135 patients with refractory metastatic colorectal cancer (mCRC) being treated with regorafenib at Samsung Medical Center, between January 2014 and January 2018. Primary tumors originating in the splenic flexure, descending colon, sigmoid colon, rectum, or proximal third of the transverse colon were defined as left-sided CRC (LC). Primary tumors originating in the appendix, cecum, ascending colon, hepatic flexure, or distal two-thirds of the transverse colon were defined as right-sided CRC (RC). Among all 135 patients, 100 (74.1%) had left sided colon cancer and 35 (25.9%) had right-sided colon cancer. No patients achieved a complete response, but four achieved a partial response, revealing a response rate (RR) of 3.0%. Thirty-seven patients had stable disease, yielding a disease control rate (DCR) of 30.4%. There was no difference in RR or DCR according to the location of the primary tumor (LC vs. RC). A significant difference in progression free survival (PFS) with regorafenib was observed between the LC and RC groups (2.6 months; 95% CI, 2.0 to 3.1 vs. 1.9 months; 95% CI, 1.6 to 2.3; P = 0.04, respectively). In a subpopulation with wild type KRAS, PFS with regorafenib was also significantly different between the LC and RC groups (2.9 months; 95% CI, 1.5 to 4.3 vs. 2.1 months; 95% CI, 0.6 to 3.6; P= 0.04). On multivariate analysis, the sidedness of the primary tumor (LC vs. RC) and the number of metastatic sites (≤1 vs. 2 & gt;) had a prognostic effect on PFS (P = 0.01 and P = 0.01, respectively). Regorafenib is a current standard treatment for CRC, but treatment outcomes may be improved if regorafenib is administered based on the appropriate biomarker. Citation Format: Sang Eun Yoon, Joon Young Hur, Su Jin Lee, Jeeyun Lee, Se Hoon Park, Joon Oh Park, Ho Yeong Lim, Won Ki Kang, Young Suk Park, Seung Tae Kim. The impact of primary tumor sidedness on the effect of regorafenib in refractory metastatic colorectal cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2912.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 1107-1107
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 1107-1107
    Abstract: We performed prospective immunohistochemical analysis of LAG3 for 430 consecutive patients with advanced gastrointestinal, genitourinary, or rare cancers between June 2012 and March 2016. Most patients (428/430, 99.5%) were evaluable for LAG3 expression by immunohistochemistry. In total, 18.5% (79/428) of the evaluated cancers expressed LAG3, including pancreatic (33.3%, 2 of 6), gastric (24.7%, 21 of 85), colorectal (23.6%, 48 of 203), melanoma (12.5%, 1 of 8), genitourinary (9.5%, 4 of 46), biliary tract (6.3%, 1 of 16), and sarcoma (5.4%, 2 of 37), but not miscellaneous (0.0%, 0 of 14) or hepatocellular (0.0%, 0 of 15) cancer. Among 149 metastatic CRC patients, there was no statistically significant difference in gender, age, primary tumor site, pathologic differentiation, KRAS and NRAS status, BRAF status, and microsatellite instability according to LAG3 status (expressed vs. non-expressed). Among 53 metastatic GC patients, LAG3 was only significantly associated with EBV status (P = .042). Our results add to the emerging literature on LAG3 expression in various cancer types and support the need for extended clinical exploration of this target for immunotherapy. Citation Format: Su Jin Lee, Sun-ju Byeon, Jeeyun Lee, Se Hoon Park, Joon Oh Park, Young Suk Park, Won Ki Kang, Ho Yeong Lim, Kyoung-Mee Kim, Seung Tae Kim. LAG3 in Solid Tumors as a Potential Novel Immunotherapy Target [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1107.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 13, No. 11 ( 2014-11-01), p. 2527-2536
    Abstract: Pazopanib is an orally bioavailable, ATP-competitive, multitargeted tyrosine kinase inhibitor mainly targeting VEGFR2 and PDGFR tyrosine kinases, but the biologic sequences of pazopanib activities beyond antiangiogenesis are poorly defined. We used a panel of 38 gastric cancer cell lines to test the efficacy of pazopanib. In a growth inhibition assay, genomic changes indicated that pazopanib had differential effects on cell growth. Treatment of the KATO-III, OCUM-2M, SNU-16, and HSC-39 gastric cancer cell lines harboring FGFR2 amplification with pazopanib resulted in marked decreases of cell survival with IC50 in ranges of 0.1 to 2.0 μmol/L, whereas the same treatment of those cell lines without FGFR2 amplification had no growth-inhibitory effects. In the ectopic FGFR2-expressing model, treatment with the indicated concentrations of pazopanib significantly inhibited cell growth and colony formation by FGFR2-expressing NIH 3T3 cells with wild-type (WT) FGFR2 and mutant FGFR2 (S252W). Pazopanib also selectively suppressed constitutive FGFR2 signaling and phosphorylation of downstream effectors. In cell-cycle analysis, FGFR2-amplified cells underwent cell-cycle arrest at the G1–S phase after pazopanib treatment, whereas there were no significant effects on cell-cycle progression in cells without FGFR2 amplification treated with pazopanib. In addition, pazopanib increased a substantial fraction of sub-G1 only in FGFR2-amplified cells. These findings show that the activation of FGFR2 signaling by amplification may be a critical mediator of cell proliferation in a small subset of gastric cancer patients and that pazopanib may provide genotype-correlated clinical benefits beyond the setting of highly vascular tumors. Mol Cancer Ther; 13(11); 2527–36. ©2014 AACR.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2062135-8
    detail.hit.zdb_id: 2063563-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 793-793
    Abstract: Background: Programmed cell death 1 (PD-1) inhibitors have shown therapeutic efficacy in metastatic gastric cancer (mGC). However, only a small portion the patients derive clinical benefits from PD-1 inhibitors, and no predictive biomarkers have been established in mGC. Inactivating mutations in serine/threonine kinase 11 (STK11), also known as liver kinase B1, are associated with immune suppression in the tumor microenvironment and poor response to PD-1 inhibitors in KRAS-mutant lung adenocarcinoma. Therefore, we hypothesized that STK11 inactivating mutations would be associated with inferior clinical response to PD-1 inhibitors in mGC. Methods: We analyzed 59 patients with mGC who had been treated with PD-1 inhibitors and whose tumors had been analyzed by targeted high-throughput sequencing. Clinicopathological characteristics, including Epstein-Barr virus positivity (EBV+), programmed death-ligand 1 (PD-L1) positivity, high microsatellite instability (MSI-H), response to PD-1 inhibitors, and survival were retrospectively evaluated. Result: STK11 mutations were identified in 30 (50.8%) patients, and were all missense mutations. Three patients (5.1%) had simultaneous STK11 gene amplification and mutation. STK11 mutations were not significantly associated with MSI-H, the number of tumor mutations, PD-L1+, EBV+, or the response to PD-1 inhibitors. Patients with STK11 mutations had prolonged overall survival (median: 19.0 vs 11.6 months, p=0.15), and progression-free survival (4.2 vs 1.9 months, p=0.06) when treated with PD-1 inhibitors, but these differences were not statistically significant. In addition, the distribution of STK11 mutations was not associated with the response to PD-1 inhibitor treatment. Patients with STK11 inactivating mutations without STK11 gene amplification had significantly prolonged progression-free survival compared to patients with intact wild type STK11 or STK11 gene amplification (4.8 vs 1.0 months, p=0.04). However, in multivariate Cox regression analysis with MSI-H, PD-L1+, EBV+, the number of mutations, and type of PD-1 inhibitor used (pembrolizumab vs nivolumab), only MSI-H and PD-L1+ were significantly associated with longer progression-free survival. Conclusions: In mGC, the presence of STK11 mutation was not predictive of the response to PD-1 inhibitors. Instead, patients with MSI-H or PD-L1+ tumors displayed superior clinical responses to PD-1 inhibitors. Citation Format: Minsuk Kwon, Kyung Kim, Suhye Choi, Jung Yong Hong, Seung Tae Kim, Se Hoon Park, Joon Oh Park, Young Suk Park, Ho Yeong Lim, Kyoung-Mee Kim, Jeeyun Lee. Association of serine/threonine kinase 11 mutations and response to programmed cell death 1 inhibitors in metastatic gastric cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 793.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 27, No. 17 ( 2021-09-01), p. 4700-4709
    Abstract: Ceralasertib is a potent and selective oral inhibitor of the serine/threonine protein kinase ataxia telangiectasia and Rad3-related (ATR) protein. Patients and Methods: Eligible patients with solid tumors, enriched for melanoma, received ceralasertib in combination with a fixed dose of paclitaxel (80 mg/m2 on D1, D8, D15) in 28-day cycles. The dose of ceralasertib was escalated to reach an MTD in a rolling 6 design. The starting dose of ceralasertib was 40 mg QD. Fifty-seven patients (33 patients with melanoma who failed prior PD1/L1 treatment) were enrolled in 7 dose cohorts ranging from 40 mg QD to 240 mg BD plus weekly paclitaxel. Results: The RP2D was established as ceralasertib 240 mg BD days 1–14 plus paclitaxel 80 mg/m2 on D1, D8, D15 every 28 days. The most common toxicities were neutropenia (n = 39, 68%), anemia (n = 25, 44%), and thrombocytopenia (n = 21, 37%). In the full analysis set of 57 patients, the overall response rate (ORR) was 22.6% (95% CI, 12.5–35.3). In 33 patients with melanoma, resistant to prior anti-PD1 therapy, the ORR was 33.3% (95% CI, 18.0–51.8). In the melanoma subset, the mPFS was 3.6 months (95% CI, 2.0–5.8), the median duration of response was 9.9 months (95% CI, 3.7–23.2), and the mOS was 7.4 months (95% CI, 5.7–11.9). Conclusions: Ceralasertib in combination with paclitaxel was well tolerated in patients with advanced malignancies and showed evidence of antitumor activity. Durable responses were observed in patients with advanced cutaneous, acral, and mucosal melanoma resistant to anti-PD1/L1 treatment. See related commentary by Ashworth, p. 4667
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 5034-5034
    Abstract: Background: Gemcitabine forms the backbone of chemotherapy and a recent study showed small but significant clinical benefits of erlotinib combined with gemcitabine in patients with pancreatic ductal adenocarcinoma. However, the high frequency of KRAS mutations in this population probably limits the benefits of an EGFR inhibitor, and this limitation seems to be similar to that observed in colon cancer and non-small cell lung cancer. We undertook this study to understand the clinical significance of KRAS mutation in pancreatic cancer patients who were treated with gemcitabine-based chemotherapy. Patients & Methods: We retrospectively studied 136 pancreatic cancer patients with available formalin-fixed paraffin-embedded tumor blocks from 2003 to 2009. KRAS mutations were analyzed by sequencing codons 12, 13 and 61. Results: In this study, 71 (52.2%) of the 136 pancreatic adenocarcinomas examined harbored a point mutation in codon 12 (N=70) and 61 (N =1) of KRAS. Observed point mutations at codon 12 in the order of frequency were: GGT-GAT (35G & gt;A; 41 of 71 patients, 57.7 %); GGT-GTT (35G & gt;T; 19 of 71 patients, 26.8%); GGT-CGT (34G & gt;C; 7 of 71 patients, 10.0%); and GGT-TGT (34G & gt;T; 3 of 71 patients, 4%). KRAS mutation was not associated with clinicopathological parameters such as age, sex, ECOG PS, smoking, tumor location, histologic differentiation, TNM staging, level of CA19-9 and first-line chemotherapy regimens. Patients with KRAS mutations showed a worse response and disease control rate (11.3 and 35.2%) than those with wild type KRAS (26.2 and 52.3%). KRAS mutation also adversely influenced survival of pancreatic cancer patients (mutant KRAS, 5.8 months [95% C.I. 5.1-6.5] vs. wild type KRAS, 8.0 months [95% C.I. 5.8-10.2] ; p=0.001) Multivariate analysis revealed good prognostic factors for overall survival as well to moderately differentiated histology (p & lt;0.001; hazard ratio (HR) = 0.437, 95% C.I. 0.301∼0.634), locally advanced disease (p & lt;0.001; HR 0.417, 95% C.I. 0.255∼0.681), response to first-line chemotherapy (p=0.003; HR 0.482, 95% C.I. 0.297∼0.780) and wild type KRAS (p=0.001; HR 0.523, 95% C.I. 0.355∼0.70). Conclusions: Our results suggest that KRAS mutation may play an important role as a biomarker for response to erlotinib treatment and in determining prognosis in pancreatic cancer patients who were treated with gemcitabine-based chemotherapy. These findings should be further explored in preclinical and upcoming prospective clinical trials to potentially allow the development of new treatment algorithms to identify patients who are most likely to respond to treatment and provide a rationale for personalized medicine in pancreatic cancer. 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 5034. doi:10.1158/1538-7445.AM2011-5034
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 10, No. 10 ( 2011-10-01), p. 1993-1999
    Abstract: Although erlotinib has become an important therapeutic option in addition to gemcitabine, the high frequency of KRAS mutations in pancreatic cancer probably limits the benefits. We retrospectively studied 136 pancreatic cancer patients with available formalin-fixed paraffin-embedded tumor blocks from 2003 to 2009 to understand the clinical significance of KRAS mutations in pancreatic cancer patients treated with gemcitabine-based chemotherapy. KRAS mutations were analyzed by sequencing codons 12, 13, and 61. In this study, 71 (52.2%) of the 136 pancreatic adenocarcinomas examined harbored a point mutation in codons 12 (n = 70) and 61 (n = 1) of KRAS. KRAS mutation was not associated with clinicopathologic parameters. Patients with KRAS mutations showed a worse response (11.3%) than those with wild-type KRAS (26.2%) and poor survival (mutant KRAS, 5.8 months vs. wild-type KRAS, 8.0 months; P = 0.001). Multivariate analysis revealed good prognostic factors for overall survival as well to moderately differentiated histology (P & lt; 0.001; HR = 0.437, 95% CI: 0.301–0.634), locally advanced disease (P & lt; 0.001; HR = 0.417, 95% CI: 0.255–0.681), response to first-line chemotherapy (P = 0.003; HR = 0.482, 95% CI: 0.297–0.780), and wild-type KRAS (P = 0.001; HR = 0.523, 95% CI: 0.355–0.770). However, the observed survival advantage is derived from the subgroup of patients treated with gemcitabine/erlotinib (9.7 vs. 5.2 months; P = 0.002), whereas no survival difference based on KRAS mutation status is obvious in the other subgroup of patients treated without erlotinib (7.0 vs. 7.0 months; P = 0.121). These results need to be further explored in upcoming prospective studies to provide a rationale for personalized medicine in pancreatic cancer. Mol Cancer Ther; 10(10); 1993–9. ©2011 AACR.
    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
    detail.hit.zdb_id: 2063563-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Molecular Cancer Therapeutics Vol. 18, No. 12_Supplement ( 2019-12-01), p. C058-C058
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 12_Supplement ( 2019-12-01), p. C058-C058
    Abstract: Vitamin C (L-Ascorbic acid, VC), an anti-oxidant has been studied as the possible potent chemotherapeutic drug in various cancers including colorectal cancer, breast cancer, ovarian cancer, prostate cancer and lung cancer. In particular, it is recently reported that VC kills efficiently colorectal cancer cells with KRAS or BRAF mutations. Give fact that pancreatic cancer has high frequency of KRAS mutation rate and is one of the most aggressive and lethal cancers with lack of effective therapy targeting the KRAS mutations, we would like to investigate whether VC effectively kills pancreatic cancer cells with various types of KRAS mutation. We first tested cytotoxicity and cell growth inhibition effect of VC on pancreatic cancer cell lines. In growth inhibition and colony formation assay, VC showed differential effect on cell growth and colony formation according to the KRAS genotypes. Treatment of the pancreatic cancer cell lines harboring KRAS G12D mutation with VC resulted in marked decrease of cell survival and colony formation, while same treatment of the cell lines with other KRAS mutations or wild type had no growth inhibitory effect. Interestingly, although there was no dramatic difference in well-known VC transporters, Glut1 expression according to the KRAS genotypes, sodium-dependent vitamin c transporter 2 (SVCT2) expression was relatively lower in KRAS G12D-mutant pancreatic cancer cells than other cells. To study possible mechanisms of VC-induced growth inhibition and cell death, we also tested whether glycolysis was inhibited or not though extracellular acidification rate (ECAR) measurement. After 1mM VC injection, ECAR decreased dramatically in KRAS G12D-mutant pancreatic cancer cells compared to the other genotypes. In cell cycle phospho-antibody array analysis, the results indicated that VC significantly increased the expression of DNA damage-associated proteins. Finally, we also found that cell death measured by apoptosis increased in KRAS G12D-mutant pancreatic cancer cells with VC through annexin V-PI staining FACS analysis. In conclusion, we showed VC selectively induces the apoptosis and cell growth inhibition in KRAS G12D-mutant pancreatic cancer cells than other KRAS mutations by inhibition of glycolysis and induction of DNA damage-associated proteins. Our findings suggest that VC is needed to further investigate as the potential novel agent with cytotoxic chemotherapies for the treatment of pancreatic cancer. Citation Format: Hye-Lim Jang, Jung Yong Hong, Seung Tae Kim, Jeeyun Lee, Se Hoon Park, Young Suk Park, Ho Yeong Lim, Won Ki Kang, Joon Oh Park. Vitamin C preferentially induces growth inhibition and cell death in KRAS G12D-mutant pancreatic cancer cells [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 C058. doi:10.1158/1535-7163.TARG-19-C058
    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
    detail.hit.zdb_id: 2063563-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4215-4215
    Abstract: Purpose We explored PD-L1 expression and analyzed its association with molecular subtypes, clinical features and other molecular markers in Korean gastric cancer (GC) patients. Experimental design As with the Asian Cancer Research Group (ACRG) GC cohort, 300 primary tumor specimens were procured from Samsung Medical Center. Tumor PD-L1 expression was measured by immunohistochemistry (IHC) using the 22C3 PD-L1 antibody pharmDx (Dako) kit. PD-L1 positivity was defined as CPS ≥1%, where CPS is PD-L1+ cells (tumor cells, macrophages, lymphocytes) over the total number of tumor cells, expressed as a percentage. ACRG and The Cancer Genome Atlas (TCGA) molecular subtypes, and other molecular and clinical features including PD-L1 mRNA expression by microarray, tumor mutation burden (TMB) by targeted sequencing, microsatellite instability (MSI) status by IHC, Epstein-Barr virus (EBV) infection by in situ hybridization, Helicobacter pylori (H. pylori) infection by microscope, ERBB2 (HER2) and EGFR overexpression by IHC, signet ring cell and Lauren classification were provided as part of the ACRG collaboration. Spearman correlation test, Wilcoxon rank-sum test, Kruskal-Wallis test were utilized to test the association with PD-L1 expression, and survival was analyzed by Kaplan-Meier method and log-rank test. Results The median age was 64 years (range, 24-86), 101 (34%) were female, and 127 (42%) had stage I/II disease. Prevalence of PD-L1 positivity was 59.3%, with significantly higher expression in stage I samples (P = 0.002). PD-L1 positivity was associated with good prognosis (P = 0.008) especially for early stage (I/II) subjects (N = 127, P = 0.007). PD-L1 mRNA was moderately correlated with protein expression (R = 0.430, P & lt; 0.001). For molecular subtype, PD-L1 expression was associated with ACRG subtype (P & lt; 0.001) with significantly higher expression in MSI subtype (P & lt; 0.001). And for TCGA subtype, the association with PD-L1 expression also existed (N = 238, P & lt; 0.001) with significantly higher expression in MSI and EBV subtype (P & lt; 0.001). H. pylori infection was also associated with higher PD-L1 expression (N = 127, P = 0.001). Signet ring cell carcinoma had significantly lower PD-L1 expression (P & lt; 0.001). PD-L1 expression was also significantly higher in ARID1A or PIK3CA mutant samples (N = 233, FDR adjusted P = 0.038). TMB was found to be weakly correlated with PD-L1 expression (N = 233, R = 0.266, P & lt; 0.001). No significant association was observed between PD-L1 expression and overexpression of ERBB2 or EGFR. Conclusion Our study provides evidence that several molecular features, especially MSI and EBV infection, are significantly associated with higher PD-L1 protein expression, which suggests that PD-L1 IHC may be an appropriate biomarker to screen GC patients for immune checkpoint inhibitors. Citation Format: Xiaoqiao Liu, Jeeyun Lee, Kyoung-Mee Kim, Seung Tae Kim, Se Hoon Park, Won Ki Kang, Razvan Cristescu, Senaka Peter. Comprehensive investigation of programmed death receptor ligand 1 (PD-L1) expression and associated molecular features in gastric cancer patients [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 4215.
    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
    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...