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)
Material
Publisher
  • American Association for Cancer Research (AACR)  (11)
Language
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2008
    In:  Cancer Research Vol. 68, No. 16 ( 2008-08-15), p. 6789-6796
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 68, No. 16 ( 2008-08-15), p. 6789-6796
    Abstract: Overexpression of mutant p53 is a common theme in human tumors, suggesting a tumor-promoting gain-of-function for mutant p53. To elucidate whether and how mutant p53 acquires its gain-of-function, mutant p53 is inducibly knocked down in the SW480 colon cancer cell line, which contains mutant p53(R273H/P309S), and the MIA PaCa-2 pancreatic cancer cell line, which contains mutant p53(R248W). We found that knockdown of mutant p53 markedly inhibits cell proliferation. In addition, knockdown of mutant p53 sensitizes tumor cells to growth suppression by various chemotherapeutic drugs. To determine whether a gene involved in cell growth and survival is regulated by mutant p53, gene expression profiling analysis was performed and showed that the expression level of Id2, a member of the inhibitor of differentiation (Id) family, was markedly increased upon knockdown of mutant p53. To confirm this, Northern blot analysis was performed and showed that the expression level of Id2 was regulated by various mutant p53s in multiple cell lines. In addition, we found that the Id2 promoter is responsive to mutant but not wild-type p53, and mutant p53 binds to the Id2 promoter. Consistent with these observations, expression of endogenous Id2 was found to be inhibited by exogenous mutant p53 in p53-null HCT116 cells. Finally, we showed that knockdown of Id2 can restore the proliferative potential of tumor cells inhibited by withdrawal of mutant p53. Together, these findings suggest that one mechanism by which mutant p53 acquires its gain-of-function is through the inhibition of Id2 expression. [Cancer Res 2008;68(16):6789–96]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2008
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2008
    In:  Cancer Research Vol. 68, No. 19 ( 2008-10-01), p. 7855-7863
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 68, No. 19 ( 2008-10-01), p. 7855-7863
    Abstract: Beclin 1 is an essential mediator of autophagy and a regulator of cell growth and cell death. We examined the effect of Beclin 1 overexpression on the action of estradiol (E2) and two antiestrogens, raloxifene and 4-hydroxytamoxifen, in estrogen receptor α (ERα)-positive MCF-7 breast cancer cells. [3H]-thymidine incorporation studies showed that Beclin 1–overexpressing cells (MCF-7.beclin) had a lower proliferative response to E2 compared with cells transfected with vector control (MCF-7.control). There was only a 35% increase in [3H] -thymidine incorporation, after 24 hours of E2 treatment of MCF-7.beclin cells compared with untreated cells, whereas this increase was 2-fold for MCF-7.control cells. E2-induced changes in the expression of early-response genes were examined by real-time quantitiative PCR. There were significant differences in the pattern of expression of E2-induced genes c-myc, c-fos, Erg-1, and Nur77 between MCF-7.beclin and MCF-7.control cells two hours after treatment. Although E2-induced growth of MCF-7.control cells was completely inhibited by 500 nmol/L raloxifene or 500 nmol/L 4-hydroxytamoxifen, these concentrations of antiestrogens had no significant effect on the growth of MCF-7.beclin cells. Confocal microscopic and coimmunoprecipitation studies showed evidence for colocalization and association of Beclin 1 and ERα. In addition, E2 caused a decrease in Akt phosphorylation in MCF-7.beclin cells, compared with a 3-fold increase in MCF-7 cells, five minutes after treatment. These results indicate that Beclin 1 can down-regulate estrogenic signaling and growth response, and contribute to the development of antiestrogen resistance. This observation might be useful to define and overcome antiestrogen resistance of breast cancer. [Cancer Res 2008;68(19):7855–63]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2008
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 4491-4491
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 4491-4491
    Abstract: p63, a p53 family protein, acts as a sequence-specific DNA-binding transcriptional factor to regulate the expression of target genes involving in various cellular processes, including growth suppression and epithelial development. RBM38, a RNA-binding protein, is a target of p53 family and is known to interact with its target mRNAs and regulate their expression via mRNA stability. Previously, we found that RBM38 negatively regulates p63 mRNA stability through binding to the AU/U-rich element in 3′ untranslated region (3′-UTR). We also found that phosphorylation of RBM38 at Ser195 alters the conformation and function of RBM38 protein. However, it is unclear whether phosphorylation of RBM38 or miR-203 alters the ability of RBM38 to regulate p63 expression. In this study, we found that in both MCF7 and HaCaT cells, wild-type RBM38 and S195A mutant, but not phosphor-mimic S195D mutant, inhibit p63 mRNA stability, suggesting that Ser195 phosphorylation abrogates the ability of RBM38 to repress the expression of p63. Importantly, we found that phosphorylation of RBM38 disrupts the binding of RBM38 to the U-rich element in p63 3′-UTR. Additionally, we found that miR-203 inhibitor increases, whereas miR203 mimic decreases p63 expression through binding to p63 3′-UTR, consistent with a previous report. Interestingly, we found that the ability of RBM38 to repress p63 expression is disrupted by miR-203 inhibitor but enhanced by miR-203 mimic. Furthermore, we found that wild-type RBM38 protein, but not phosphor-mimic S195D mutant, physically interacts with Ago2, indicating that RBM38 may inhibit p63 mRNA stability by modulating Ago2-miR-203 complex to bind to the U-rich element in p63 3′-UTR. Together, we uncovered a novel mechanism by which RBM38 modulates p63 expression via miR-203. Note: This abstract was not presented at the meeting. Citation Format: YANHONG ZHANG, WENSHENG YAN, JIN ZHANG, XINBIN CHEN. Modulation of p63 expression by phosphorylation of RBM38 at S195 via miR-203 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4491. doi:10.1158/1538-7445.AM2017-4491
    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 ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 14 ( 2019-07-15), p. 3714-3724
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 14 ( 2019-07-15), p. 3714-3724
    Abstract: Platinum-based anticancer drugs are widely used as a first-line drug for cancers, such as non–small cell lung carcinoma (NSCLC) and bladder cancer. However, the efficacy is limited due to intrinsic or acquired resistance to these drugs. DNA polymerase eta (PolH, Polη) belongs to the Y-family of DNA polymerases and mediates DNA translesion synthesis, a major mechanism for DNA damage tolerance. Here, we showed that a high level of PolH is associated with cisplatin resistance in lung and bladder cancer. Consistent with this, loss of PolH markedly attenuates cisplatin resistance in both cisplatin-sensitive and cisplatin-resistant lung cancer cells. Interestingly, we found that due to the presence of multiple polyadenylation sites, alternative polyadenylation (APA) produces three major PolH transcripts with various lengths of 3′untranslated region (3′UTR; 427-/2516-/6245-nt). We showed that the short PolH transcript with 427-nt 3′UTR is responsible for high expression of PolH in various cisplatin-resistant lung and bladder cancer cell lines. Importantly, loss of the short PolH transcript significantly sensitizes cancer cells to cisplatin treatment. Moreover, we found that miR-619 selectively inhibits the ability of the long PolH transcript with 6245-nt 3′UTR to produce PolH protein and, subsequently, PolH-dependent cell growth. Together, our data suggest that PolH expression is controlled by APA and that the short PolH transcript produced by APA can escape miR-619–mediated repression and, subsequently, confers PolH-mediated cisplatin resistance. Significance: A short PolH transcript produced by alternative polyadenylation escapes repression by miR-619 and confers resistance to cisplatin.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2007
    In:  Cancer Research Vol. 67, No. 19 ( 2007-10-01), p. 9117-9124
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 67, No. 19 ( 2007-10-01), p. 9117-9124
    Abstract: p53 tumor suppressor and its family members, p63 and p73, are known to play a role in the survival of cells exposed to stress signals. As a transcription factor, the p53 family proteins induce a plethora of target genes that mediate their functions in the cell cycle, apoptosis, and other biological activities. However, the mechanism by which the p53 family proteins regulate their cell survival functions is still not clear. Here, we showed that bone morphogenetic protein 7 (BMP7) is a novel target gene regulated by the p53 family and mediates the cell survival function of the basal physiologically relevant level of p53. Specifically, we found that knockdown of BMP7 markedly inhibits the proliferation of p53-deficient, but not p21-knockdown, breast cancer cells compared with the ones with wild-type p53. In addition, we found that inhibitor of differentiation or DNA binding 2 (Id2), a transcription factor implicated for cell survival, is regulated by the BMP7 and p53 pathways. Interestingly, whereas a functional BMP7 or p53 pathway is sufficient to maintain the basal level of Id2 expression, loss of both pathways abrogates Id2 expression. Furthermore, we showed that overexpression of Id2 can restore p53-deficient cells to survive in the absence of BMP7. As a result, we identified a previously unrecognized role for BMP7 in the maintenance of cell survival for p53-deficient cells, at least in part, through Id2. Together, we hypothesize that breast cancer patients with mutant p53 might benefit from targeted repression of BMP7 expression and/or targeted inhibition of the BMP7 pathway. [Cancer Res 2007;67(19):9117–24]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2007
    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. 4594-4594
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 4594-4594
    Abstract: Table of Contents • Estrogen mimetic effect of a polyamine analogue in MCF-7 breast cancer cells Estrogen mimetic effect of a polyamine analogue in MCF-7 breast cancer cells Bis(ethyl) polyamine analogues have been synthesized and tested as chemotherapeutic agents for different subsets of cancer. 1,15-bis(ethylamino)4,8,12-triazapentadecane (BE-3-3-3-3) is a polyamine analogue with therapeutic potential for breast cancer. We examined the effect of BE-3-3-3-3 on estrogen receptor α (ERα) positive MCF-7 cells in the presence and absence of 4 nM estradiol (E2). In the presence of E2, a concentration-dependent decrease in DNA synthesis was observed using [3H]-thymidine incorporation assay. In the absence of E2, low concentrations (1.25 to 5 µM) of BE-3-3-3-3 increased [3H] -thymidine incorporation at 24 and 48 hours of treatment. However, growth inhibition was observed by 96 hours of treatment. In order to elucidate the mechanism of analogue action, we examined the expression of two E2 sensitive oncogenes, c-myc and c-fos, using real-time quantitative polymerase chain reaction (qPCR) method. Treatment with BE-3-3-3-3 for 2 hours induced the expression of c-myc (∼2.4-fold) and c-fos (∼4.5-fold) compared to untreated control. BE-3-3-3-3's estrogen-mimetic action was also evidenced by the increased occupancy of the pS2/Tff1 gene promoter by ERα and its co-activator, steroid receptor co-activator 3 (SRC-3), as assessed by chromatin immunoprecipitation (ChIP) assay. Confocal microscopic visualization of BE-3-3-3-3- treated cells revealed co-localization of ERα in a manner similar to that induced by E2. Western blotting analysis showed that delayed growth inhibition in the absence of E2 was associated with the induction of autophagy, as evidenced by elevated levels of autophagy-related proteins, Beclin 1 and MAP LC3 II. Electron microscopic investigation demonstrated the presence of autophagic vacuoles in analogue-treated cells. Additionally, BE-3-3-3-3 inhibited polyamine synthesis and induced catabolism, resulting in significantly decreased levels of putrescine, spermidine and spermine. Analogue treatment reduced the activity of a key enzyme in the polyamine biosynthetic pathway, ornithine decarboxylase (ODC), by ∼ 23-fold after 24 hours, and stimulated the catabolic activity of spermidine/spermine N1-acetyltransferase (SSAT) by 180-fold after 48 hours. These results provide evidence of BE-3-3-3-3's ability to modulate gene expression and stimulate proliferation of breast cancer cells under conditions of low E2 availability. Our findings may help to target this compound toward appropriate subsets of breast cancer patients. In addition, our results indicate a novel pathway for the action of bis(ethyl) polyamine analogues in the balance of cell growth and autophagy. 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 4594.
    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. 13, No. 7 ( 2014-07-01), p. 1860-1872
    Abstract: Although silybin, a natural flavonolignan, has been shown to exhibit potent antitumor activities against various types of cancers, including lung cancer, the molecular mechanisms behind these activities remain unclear. Silent information regulator 1 (SIRT1) is a conserved NAD+-dependent deacetylase that has been implicated in the modulation of transcriptional silencing and cell survival. Furthermore, it plays a key role in carcinogenesis through the deacetylation of important regulatory proteins, including p53. In this study, we investigated the antitumor activity of silybin towards human lung adenocarcinoma cells in vitro and in vivo and explored the role of the SIRT1 signaling pathway in this process. Silybin treatment resulted in a dose- and time-dependent decrease in lung adenocarcinoma A549 cell viability. In addition, silybin exhibited strong antitumor activity illustrated by reductions in tumor cell adhesion, migratory capability, and glutathione levels and by increased apoptotic indices and reactive oxygen species levels. Silybin treatment also downregulated SIRT1 and upregulated p53 acetylation. SIRT1 siRNA (in vitro) or cambinol (a known SIRT1 inhibitor used for in vivo studies) further enhanced the antitumor activity of silybin. In summary, silybin is a potent inhibitor of lung adenocarcinoma cell growth that interferes with SIRT1 signaling, and this inhibition is a novel mechanism of silybin action that may be used for therapeutic intervention in lung adenocarcinoma treatment. Mol Cancer Ther; 13(7); 1860–72. ©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
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 13 ( 2011-07-01), p. 4585-4597
    Abstract: Trastuzumab shows remarkable efficacy in treatment of ErbB2-positive breast cancers when used alone or in combination with other chemotherapeutics. However, acquired resistance develops in most treated patients, necessitating alternate treatment strategies. Increased aerobic glycolysis is a hallmark of cancer and inhibition of glycolysis may offer a promising strategy to preferentially kill cancer cells. In this study, we investigated the antitumor effects of trastuzumab in combination with glycolysis inhibitors in ErbB2-positive breast cancer. We found that trastuzumab inhibits glycolysis via downregulation of heat shock factor 1 (HSF1) and lactate dehydrogenase A (LDH-A) in ErbB2-positive cancer cells, resulting in tumor growth inhibition. Moreover, increased glycolysis via HSF1 and LDH-A contributes to trastuzumab resistance. Importantly, we found that combining trastuzumab with glycolysis inhibition synergistically inhibited trastuzumab-sensitive and -resistant breast cancers in vitro and in vivo, due to more efficient inhibition of glycolysis. Taken together, our findings show how glycolysis inhibition can dramatically enhance the therapeutic efficacy of trastuzumab in ErbB2-positive breast cancers, potentially useful as a strategy to overcome trastuzumab resistance. Cancer Res; 71(13); 4585–97. ©2011 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 675-675
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 675-675
    Abstract: The nerve injury-induced protein 1 (Ninjurin1, Ninj1), a homophilic adhesion molecule, is involved in nerve regeneration, inflammation and vascular regression. Ninj1 is found to be overexpressed in human cancers including hepatocellular carcinoma and acute lymphoblastic leukemia. However, the role of Ninj1 in tumorigenesis has not been elucidated. Recently, we found that Ninj1 is transcriptionally regulated by p53, which in turn regulates p53 expression through mRNA translation. Thus, the mutual regulation between p53 and Ninj1 represents a novel loop in the p53 pathway. To examine the biological significance of the p53-Ninj1 loop, we generated and monitored 4 cohorts of mice: (i) WT, (ii) Ninj1-het, (iii) p53-null, (iv) Ninj1-het; p53-null. We found that Ninj1-het mice were highly prone to skin lesions. We also found that Ninj1 deficiency shortened the life-span but decreased the incidence of sarcoma in mice without p53. Additionally, we found that Ninj1-het; p53-null mice were prone to hydrocephalus and kidney abnormality. To examine the association of Ninj1 with mutant p53, we generated and characterized Ninj1+/-; p53 R270H/- and p53 R270H/- mice. We found that Ninj1 deficiency shortened the life-span and promoted the frequency and aggressiveness of tumors in p53 R270H/- mice. Together, we conclude that Ninj1 differently regulates the tumor susceptibility in a p53-dependent manner in vivo and that the Ninj1-p53 loop plays a pivotal role in tumor suppression and longevity. Citation Format: Hee Jung Yang, Seong Jun Cho, Jin Zhang, Wensheng Yan, Xinbin Chen. Ninjurin1, a target of p53, modulates p53-dependent tumor suppression in vivo. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 675.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 8_Supplement ( 2023-04-14), p. CT075-CT075
    Abstract: Background: The ORIENT-15 study (NCT03748134) evaluated sintilimab (anti-PD-1 antibody) plus chemotherapy (Sin+Chemo) versus placebo plus chemotherapy (Chemo) as first-line (1L) treatment of unresectable locally advanced, recurrent, or metastatic esophageal squamous cell carcinoma (ESCC). At the prespecified interim analysis, this study met the primary endpoints of overall survival (OS) in all patients (pts) (hazard ratio [HR] 0.63; 95% confidence interval [CI] 0.51-0.78; P & lt;0.001) and the pts with PD-L1 combined positive score (CPS) ≥10 (HR 0.64; 95% CI 0.48-0.85; P=0.002) (Lu, et al. BMJ 2022). Here we report the updated OS results with an extended follow-up time. Methods: Eligible pts were randomized 1:1 to receive sintilimab/placebo (3 mg/kg in pts weighing & lt;60 kg or 200 mg in pts weighing ≥60 kg, IV Q3W) for up to 24 months plus chemo (paclitaxel 175 mg/m2 Q3W plus cisplatin 75 mg/m2 Q3W as TP regimen, or cisplatin 75 mg/m2 Q3W plus 5-FU 800 mg/m2 on days 1-5 Q3W as CF regimen). Stratification factors were PD-L1 expression (tumor proportion score & lt;10% or ≥10%), ECOG PS (0 vs 1), liver metastasis (presence vs absence), and chemo regimen (TP vs CF). The primary endpoints were OS in the pts with PD-L1 CPS ≥10 and all pts. Results: Overall, 690 pts (median age 63.0; 85.5% male; 93.3% Asian and 4.9% White; 24.3% liver metastasis; 90.7% TP regimen) were randomized and received Sin+Chemo (341 pts) or Chemo (349 pts). As of data cutoff date (Aug 28, 2022), the median follow-up was 32.2 months (interquartile range 28.0-35.8); 231 OS events were observed in Sin+Chemo group and 278 in Chemo group. Sin+Chemo continued to demonstrate an OS benefit vs Chemo in all pts (median OS 17.4 [95% CI 16.0-19.8] vs 12.8 [95% CI 11.3-14.5] months; HR 0.661 [95% CI: 0.554-0.788]; P & lt;0.0001), and the pts with PD-L1 CPS ≥10 (18.4 [95% CI 16.2-24.6] vs 14.5 [95% CI 11.7-16.4] months; HR 0.635 [95% CI 0.503-0.803]; P=0.0001). Estimated OS rates at 12 and 24 months for Sin+Chemo vs Chemo in all pts were 64.0% vs 53.5% and 41.4% vs 22.9%, respectively. Subgroup analyses of OS in all pts were generally consistent with the previous report, showing homogeneity in OS outcomes. The CTCAE grade ≥3 treatment-related adverse events occurred in 60.7% pts in Sin+Chemo group and 56.2% pts in Chemo group. No new or unexpected safety signals were observed. Conclusions: Sin+Chemo continued to demonstrate significant OS benefits in advanced ESCC in both overall and PD-L1 CPS ≥10 populations with an acceptable safety profile over time. These data further support the use of Sin+Chemo as a standard of care for 1L treatment in these patients. Citation Format: Zhihao Lu, Junye Wang, Yongqian Shu, Li Kong, Buhai Wang, Lei Yang, Guochun Cao, Guogui Sun, Yinghua Ji, Hu Liu, Tongjian Cui, Wensheng Qiu, Aziz Zaanan, Roberto Pazo Cid, Hui Zhou, Xing Sun, Yan Wang, Yuling Chen, Haoyuan Li, Lishi Zhang, Lin Shen. Updated overall survival outcomes from a randomized, double-blind phase III study of sintilimab versus placebo in combination with chemotherapy as first-line treatment for advanced esophageal squamous cell carcinoma (ORIENT-15) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT075.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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...