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)  (20)
  • 1
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 27, No. 10 ( 2021-05-15), p. 2734-2741
    Abstract: This surgical window of opportunity (window) study assessed the short-term effect of medroxyprogesterone acetate (MPA) alone versus MPA plus the histone deacetylase (HDAC) inhibitor entinostat on regulation of progesterone receptor (PR) in women with newly diagnosed endometrioid endometrial adenocarcinoma. Patients and Methods: This multisite, randomized, open-label surgical window study treated women intramuscularly on day 1 with 400 mg MPA. Entinostat given 5 mg by mouth on days 1, 8, and 15 was randomly assigned with equal probability. Surgery followed on days 21–24. Pretreatment and posttreatment tissue was assessed for PR H-scores, Ki-67 levels, and histologic response. Results: Fifty patients were accrued in 4 months; 22 and 20 participants had PR evaluable pretreatment and posttreatment slides in the MPA and MPA/entinostat arms, respectively. Median posttreatment PR H-scores were significantly lower than pretreatment H-scores in both arms but did not differ significantly (MPA: 247 vs. 27, MPA/entinostat 260 vs. 23, respectively, P = 0.87). Decreased Ki-67 was shown in 90% treated with MPA/entinostat compared with 68% treated with MPA alone (P = 0.13). Median PR H-score decreases were larger when Ki-67 was decreased (208) versus not decreased (45). The decrease in PR pretreatment versus posttreatment was associated with loss of Ki-67 nuclear staining, consistent with reduced cellular proliferation (P & lt; 0.008). Conclusions: This surgical window trial rapidly accrued in a multisite setting and evaluated PR as its primary endpoint and Ki-67 as secondary endpoint. Despite no immediate effect of entinostat on PR in this short-term study, lessons learned can inform future window and treatment trials.
    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 ...
  • 2
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 4, No. 1 ( 2014-01-01), p. 80-93
    Abstract: BRAF inhibitors elicit rapid antitumor responses in the majority of patients with BRAFV600-mutant melanoma, but acquired drug resistance is almost universal. We sought to identify the core resistance pathways and the extent of tumor heterogeneity during disease progression. We show that mitogen-activated protein kinase reactivation mechanisms were detected among 70% of disease-progressive tissues, with RAS mutations, mutant BRAF amplification, and alternative splicing being most common. We also detected PI3K–PTEN–AKT–upregulating genetic alterations among 22% of progressive melanomas. Distinct molecular lesions in both core drug escape pathways were commonly detected concurrently in the same tumor or among multiple tumors from the same patient. Beyond harboring extensively heterogeneous resistance mechanisms, melanoma regrowth emerging from BRAF inhibitor selection displayed branched evolution marked by altered mutational spectra/signatures and increased fitness. Thus, melanoma genomic heterogeneity contributes significantly to BRAF inhibitor treatment failure, implying upfront, cotargeting of two core pathways as an essential strategy for durable responses. Significance: This study provides critical insights into how human BRAF-mutant melanoma, a malignancy with marked mutational burden, escapes from BRAF inhibitors. Understanding the core resistance pathways as well as tumor heterogeneity, fitness, and mutational patterns, which emerge under drug selection, lays a foundation to rationalize clinical studies and investigate mechanisms of disease progression. Cancer Discov; 4(1); 80–93. ©2013 AACR. See related commentary by Solit and Rosen, p. 27 This article is highlighted in the In This Issue feature, p. 1
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 3, No. 1 ( 2013-01-01), p. 96-111
    Abstract: Persistent signaling by the oncogenic EGF receptor (EGFR) is a major source of cancer resistance to EGFR targeting. We established that inactivation of 2 sterol biosynthesis pathway genes, SC4MOL (sterol C4-methyl oxidase–like) and its partner, NSDHL (NADP-dependent steroid dehydrogenase–like), sensitized tumor cells to EGFR inhibitors. Bioinformatics modeling of interactions for the sterol pathway genes in eukaryotes allowed us to hypothesize and then extensively validate an unexpected role for SC4MOL and NSDHL in controlling the signaling, vesicular trafficking, and degradation of EGFR and its dimerization partners, ERBB2 and ERBB3. Metabolic block upstream of SC4MOL with ketoconazole or CYP51A1 siRNA rescued cancer cell viability and EGFR degradation. Inactivation of SC4MOL markedly sensitized A431 xenografts to cetuximab, a therapeutic anti-EGFR antibody. Analysis of Nsdhl-deficient Bpa1H/+ mice confirmed dramatic and selective loss of internalized platelet-derived growth factor receptor in fibroblasts, and reduced activation of EGFR and its effectors in regions of skin lacking NSDHL. Significance: This work identifies a critical role for SC4MOL and NSDHL in the regulation of EGFR signaling and endocytic trafficking and suggests novel strategies to increase the potency of EGFR antagonists in tumors. Cancer Discov; 3(1); 96–111. ©2012 AACR. This article is highlighted in the In This Issue feature, p. 1
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 1, No. 6 ( 2013-12-01), p. 373-377
    Abstract: Immune checkpoint inhibitors such as ipilimumab and targeted BRAF inhibitors have dramatically altered the landscape of melanoma therapeutics over the past few years. Agents targeting the programmed cell death-1/ligand (PD-1/PD-L1) axis are now being developed and seem to be highly active clinically with favorable toxicity profiles. We report on two patients with BRAF V600E–mutant melanoma who were treated with anti-PD-1 agents as first-line therapy without significant toxicity, followed by vemurafenib at disease progression. Both patients developed severe hypersensitivity drug eruptions with multiorgan injury early in their BRAF inhibitor treatment course. One patient subsequently developed acute inflammatory demyelinating polyneuropathy, and the other developed anaphylaxis upon low-dose vemurafenib rechallenge. Further investigation of the immune response during combination or sequences of melanoma therapeutics is warranted. Furthermore, clinicians should maintain a high index of suspicion for these toxicities when vemurafenib is administered following an anti-PD-1 agent. Cancer Immunol Res; 1(6); 373–7. ©2013 AACR.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 20, No. 12_Supplement ( 2021-12-01), p. P02-02-P02-02
    Abstract: INTRODUCTION: Oncogenic FGFR2 alterations (fusions/rearrangements, amplifications, mutations) are key drivers in cholangiocarcinoma (CCA) and multiple solid tumors. Current pan-FGFR inhibitor (FGFRi) therapy is limited by off-isoform toxicity and acquired FGFR2 kinase domain resistance mutations. RLY-4008 is a highly selective and potent oral inhibitor designed to target both FGFR2 driver and resistance mutations. We initiated a first-in-human study in advanced solid tumors patients (pts) to define the safety, pharmacokinetics (PK) and efficacy of RLY-4008 (NCT04526106). METHODS: Adult pts received RLY-4008 QD or BID on a 4-week cycle following a BOIN escalation design. Adverse events (AEs), PK, ctDNA and anti-tumor activity (RECIST 1.1) were assessed. RESULTS: As of 16AUG21, 45 pts (35 CCA; 10 other) have been treated with RLY-4008 at total daily doses of 30-200 mg (18 pts BID; 27 pts QD). 44 pts had oncogenic FGFR2 alterations (26 fusions/13 mutations/5 amplifications). The median number of prior anti-neoplastic therapies was 3 (range 1-15). 94% (33/35) of CCA pts had prior chemotherapy and 69% (24/35) had prior FGFRi. 56% (9/16) CCA pts with prior FGFRi and evaluable ctDNA had ≥1 FGFR2 resistance mutation at baseline, most commonly at positions 549 (8/9), 617 (3/9), or 564 (2/9). RLY-4008 had rapid absorption (Tmax 1-7h), half-life to support QD dosing (18-34 h), dose-dependent exposure (AUC; Cmax) and predicted FGFR2 occupancy & gt;85% across dose levels. The MTD has not been defined, and QD dose exploration continues to select the optimal biologically efficacious dose. AEs occurring in & gt;20% of pts include stomatitis (49%), palmar-plantar erythrodysesthesia (PPE, 38%), dry mouth (29%), and nail toxicities (22%), majority of which were ≤Gr 2. 6 pts had Gr 1-2 retinopathy, which resolved in all cases. 5 AEs were considered dose limiting toxicities: 4 in BID (rash/PPE/mucositis/hyperbilirubinemia) and 1 in QD (retinopathy). No Gr 4/5 drug-related AEs were seen. 25 pts remain on treatment (range 1-37 weeks). RLY-4008 showed broad anti-tumor activity across dose levels and FGFR2 alterations with radiographic tumor reductions of ≥10% in 59% pts (19/32; -11% to -83%). Activity was seen in FGFRi-naïve, FGFR2-fusion+ CCA with PRs in 50% of pts (3/6, 2 confirmed and 1 pending confirmation; -56% to -83%). Activity was also seen in FGFRi pre-treated FGFR2-fusion+ CCA pts (N=16) with 16 SD, including 9 pts with tumor reduction ≥10% (from -12% to -35%). Of the FGFRi pre-treated FGFR2-fusion+ CCA patients with detectable FGFR2 resistance mutations in ctDNA at baseline, 78% (7/9) were undetectable at C2D1. CONCLUSION: RLY-4008 demonstrates promising safety, tolerability, and clinical activity in FGFR2-altered solid tumor pts, including those who progressed on prior FGFRi therapy. Consistent with the FGFR2-selective mechanism, minimal off-isoform toxicity (FGFR1-hyperphosphatemia; FGFR4-diarrhea) was seen. These encouraging data validate selective targeting of FGFR2 and suggest that RLY-4008 has potential to overcome resistance to FGFRi. Citation Format: Lipika Goyal, Mitesh Borad, Vivek Subbiah, Amit Mahipal, Suneel Kamath, Kabir Mody, Robin Katie Kelley, Richard Kim, Vaibhav Sahai, Anthony El-Khoueiry, Efrat Dotan, Oleg Schmidt-Kittler, Jinshan Shen, Kai Yu Jen, Alicia Deary, Wei Guo, Mahesh Padval, Cori Ann J. Sherwin, Charles Ferte, Beni Wolf, Alison M. Schram. First results of RLY-4008, a potent and highly selective FGFR2 inhibitor in a first-in-human study in patients with FGFR2-altered cholangiocarcinoma and multiple solid tumors [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2021 Oct 7-10. Philadelphia (PA): AACR; Mol Cancer Ther 2021;20(12 Suppl):Abstract nr P02-02.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 16, No. 2 ( 2010-01-15), p. 681-690
    Abstract: Purpose: To identify molecular markers of pathologic response to neoadjuvant paclitaxel/radiation treatment, protein and gene expression profiling were done on pretreatment biopsies. Experimental Design: Patients with high-risk, operable breast cancer were treated with three cycles of paclitaxel followed by concurrent paclitaxel/radiation. Tumor tissue from pretreatment biopsies was obtained from 19 of the 38 patients enrolled in the study. Protein and gene expression profiling were done on serial sections of the biopsies from patients that achieved a pathologic complete response (pCR) and compared to those with residual disease, non-pCR (NR). Results: Proteomic and validation immunohistochemical analyses revealed that α-defensins (DEFA) were overexpressed in tumors from patients with a pCR. Gene expression analysis revealed that MAP2, a microtubule-associated protein, had significantly higher levels of expression in patients achieving a pCR. Elevation of MAP2 in breast cancer cell lines led to increased paclitaxel sensitivity. Furthermore, expression of genes that are associated with the basal-like, triple-negative phenotype were enriched in tumors from patients with a pCR. Analysis of a larger panel of tumors from patients receiving presurgical taxane-based treatment showed that DEFA and MAP2 expression as well as histologic features of inflammation were all statistically associated with response to therapy at the time of surgery. Conclusion: We show the utility of molecular profiling of pretreatment biopsies to discover markers of response. Our results suggest the potential use of immune signaling molecules such as DEFA as well as MAP2, a microtubule-associated protein, as tumor markers that associate with response to neoadjuvant taxane–based therapy. Clin Cancer Res; 16(2); 681–90
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 4942-4942
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 4942-4942
    Abstract: Distant tumor metastases and their confounding symptoms are often responsible for reduced patient quality-of-life and decreased overall survival. The main route of metastasis for many solid tumors is the lymphatic vasculature. The mechanisms of metastatic spread of tumor cells through the lymphatic vessels remain poorly understood. We have developed a novel transgenic mouse model and customized real-time intravital microscopy platforms to study the migration of tumor cells into lymphatic vessels in live animals over the course of several days. TdTomato fluorescent protein was expressed in lymphatic endothelial cells using an inductive CRE recombinase approach. This transgenic system coupled with the use of fluorescent stereomicroscopy techniques allowed us to visualize the cutaneous lymphatic vessel network with remarkable anatomic detail in living mice. We tracked the fate of CFSE-labeled B16 tumor cells administered cutaneously using two-color intravital fluorescence microscopy. This strategy allowed us to simultaneously visualize the lymphatic vessel network (red) and the B16 tumor cells (green) at single-cell resolution. We clearly visualized and documented several key behaviors of metastasizing tumor cells including single-cell intravasation, movement of tumor cells within vessels, clearing from vessels, and the accumulation of tumor cells in regional lymph nodes. These events occurred at a low frequency over the course of days in conditions of tissue homeostasis. Current studies are designed to define the anatomic site of tumor intravasation into lymphatic vessels and investigate how the inflamed tumor microenvironment affects tumor migration into the lymphatic vasculature. Citation Format: Darci M. Fink, Alicia L. Connor, Philip M. Kelley, Richard M. Tempero, Michael A. Hollingsworth. Metastatic tumor migration in lymphatic vessels revealed by real-time intravital imaging. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4942. doi:10.1158/1538-7445.AM2014-4942
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 2145-2145
    Abstract: In the phase 3 CheckMate 459 study (NCT02576509), first-line nivolumab (NIVO) treatment demonstrated higher overall response rate (ORR) and a favorable safety profile, but no significant improvement in overall survival (OS) compared with sorafenib (SORA) in patients with advanced hepatocellular carcinoma (HCC). Here, we report exploratory biomarker analyses from the trial with a potential to identify patients who may benefit from NIVO. Archival or fresh tumor samples were collected before treatment and subjected to whole exome sequencing (WES) (400/730: 55%) for genetic alterations including high microsatellite instability (MSI-H) and WNT/beta-catenin pathway components, as well as whole transcriptome RNA sequencing (469/730: 64%) for analysis of individual genes, inflammation signatures and Gene Set Enrichment Analysis (GSEA). Associations between biomarkers and best overall response (BOR), progression free survival (PFS) and OS were evaluated at a minimum follow-up of 33·6 months. RNA sequencing analysis demonstrated that high Gajewski inflammation gene signature score of ≥0.47 (30% of tumors) was associated with an improvement in BOR (p=0.0043), PFS (p=0.00065) and OS (p=0.0073) within the NIVO arm only. Upon comparison of NIVO versus SORA, only patients whose tumors were inflammation high demonstrated NIVO benefit for BOR (p=0.017), PFS (p=0.0063) and OS (p=0.037). Responses to NIVO were associated with higher expression of CD8A, CD8B, PDCD1 (PD-1) and other inflammation-associated genes within the tumor (false discovery rate, FDR & lt;0.01). GSEA of 50 hallmark gene sets for ORR showed that responders to NIVO and SORA were enriched in similar gene sets such as inflammatory response, interferon alpha/gamma response, E2F targets and G2M checkpoint (FDR & lt;0.01). In contrast, GSEA for OS showed inflammatory response and IL6-JAK-STAT3 signaling gene sets were associated with benefit from NIVO, but shorter OS for SORA (FDR & lt;0.01). WES analysis demonstrated that the prevalence of MSI-H was low (12/400: 3%) and insufficient to confirm relationship to clinical outcomes (no radiographic responses observed in 9 NIVO- and 3 SORA-treated patients). Alterations within beta-catenin gene were not associated with survival outcomes for NIVO. Overall, patients with advanced HCC whose tumors had high Gajewski inflammation gene signature demonstrated improved outcomes with NIVO versus SORA. NIVO responders had higher expression of individual inflammation-associated genes. Expression of inflammatory response and IL6-JAK-STAT3 signaling gene sets showed association with longer OS for NIVO, but not SORA. These exploratory analyses support further studies of tumor inflammation-related biomarkers to identify patients most likely to benefit from PD-1 inhibition in advanced HCC. Citation Format: Jaclyn Neely, Jin Yao, Masatoshi Kudo, Richard S. Finn, Bruno Sangro, Ignacio Melero, Anthony El-Khoueiry, Marina Tschaika, Damir Begic, Ashwin Sama, Parul Doshi, Thomas Yau, Robin Kate Kelley. Genomic and transcriptomic analyses related to the clinical efficacy of first-line nivolumab in advanced hepatocellular carcinoma from the phase 3 CheckMate 459 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 2145.
    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 ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 5098-5098
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 5098-5098
    Abstract: Epidermal growth factor receptor (EGFR) has been exploited as a target for blockade in cancer therapy, based on the known central role of EGFR signaling in promoting cancer cell growth. We have identified a novel mechanism for influencing EGFR activity that involves the sterol biosynthesis pathway enzyme sterol C4-methyl oxidase-like (SC4MOL). SC4MOL, in conjunction with other members of the ergosome complex (erg26/NSDHL; erg27/HSD17B7), catalyzes a non-redundant, irreversible step of demethylation at C4 of 14-demethylated lanosterol and dihydrolanosterol to produce zymosterol and 8(9)-cholestenol, respectively. Depletion of SC4MOL with siRNA caused accumulation of methylsterol intermediaries; inhibition of the key EGFR effectors, ERK and AKT; and a 3-fold increase in apoptosis specifically in the presence of EGFR antagonists. Metabolic block upstream of SC4MOL with ketoconazole or CYP51A1 siRNA eliminated methylsterols and completely reversed the effect of SC4MOL depletion, while added cholesterol did not rescue cancer cells from accelerated apoptosis upon concurrent SC4MOL and EGFR blockade. These results indicate that accumulation of specific cholesterol precursors enhances efficacy of anti-EGFR drugs. Further, we found that SC4MOL and the other ergosome components are transcriptionally regulated by EGFR signaling, suggesting involvement of C4 methylsterols in physiological activation of the EGFR signaling pathway. A metabolic effect of sterol intermediaries on EGFR signaling has not been previously characterized. Such an effect could be exploited in cancer therapeutically by employing already existing drugs such as synthetic 4,4-dimethylsterols currently in use for in vitro fertilization. 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 5098.
    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 ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. CT283-CT283
    Abstract: Background: Biliary tract cancer (BTC), including intra- or extrahepatic cholangiocarcinoma (CCA) and gallbladder cancer, is an aggressive malignancy with limited treatment options. Most patients (pts) present with advanced or unresectable disease, for which the current first-line standard of care is gemcitabine with cisplatin. Prognosis for these pts is poor, with median survival of approximately 12 months. Pembrolizumab (pembro) has improved outcomes when combined with platinum-based chemotherapy in other tumor types (eg, gastric/GEJ, NSCLC). The randomized, placebo-controlled, double-blind, phase 3 KEYNOTE-966 trial (NCT04003636) will be conducted to evaluate pembro plus gemcitabine and cisplatin compared with placebo plus gemcitabine and cisplatin in pts with metastatic or unresectable locally advanced BTC. Methods: Pts will be randomly assigned 1:1 to receive pembro 200 mg or placebo IV every 3 weeks plus gemcitabine 1000 mg/m2 IV and cisplatin 25 mg/m2 IV on days 1 and 8 of every 3-week cycle. Cisplatin will be given for a maximum of 8 cycles. Treatment with pembro/placebo will continue for up to 35 administrations or until disease progression, unacceptable toxicity, or withdrawal from the study. Gemcitabine will be continued until disease progression, unacceptable toxicity, or withdrawal from the study. Pts will be stratified by region (Asia, non-Asia), stage (locally advanced, metastatic), and site of origin (gallbladder, intrahepatic CCA, or extrahepatic CCA). Key eligibility criteria are age ≥18 years; histologically confirmed metastatic or unresectable locally advanced BTC, measurable by RECIST v1.1; no prior systemic therapy for advanced BTC; Eastern Cooperative Oncology Group performance status of 0 or 1; and tumor sample adequate for biomarker analyses. Pts with past or ongoing hepatitis C or controlled hepatitis B virus infection are eligible per protocol-defined criteria. During the initial stage, response will be assessed by imaging every 6 weeks until week 54 and every 12 weeks thereafter. Safety, assessed by CTCAE v.5.0, will be monitored throughout the study and for 30 days after treatment end or 90 days for serious adverse events. The primary end points are progression-free survival (as assessed by blinded independent central review) and overall survival (OS). Secondary end points are objective response rate, duration of response, and safety. Additional analyses will include disease control rate, health-related quality of life (by EORTC QLQ-C30, EuroQol EQ-5D, and EORTC QLQ-BIL21), and molecular and genetic biomarkers. The study opened to recruitment on September 24, 2019. Recruitment is underway in 17 countries and will continue until approximately 788 pts are enrolled. Citation Format: Richard S. Finn, Robin K. Kelley, Junji Furuse, Julien Edeline, Zhenggang Ren, Shu-Chih Su, Usha Malhotra, Abby B. Siegel, Juan W. Valle. KEYNOTE-966: A randomized, double-blind, placebo-controlled, phase 3 study of pembrolizumab in combination with gemcitabine and cisplatin for the treatment of advanced biliary tract carcinoma [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 CT283.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...