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)  (8)
Type of Medium
Publisher
  • American Association for Cancer Research (AACR)  (8)
Language
Subjects(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 1937-1937
    Abstract: Small cell lung cancer (SCLC) accounts for approximately 15% of all lung cancers, leading to ~30,000 deaths each year in the United States. SCLC patients often present with metastasis at time of diagnosis, excluding surgery as a treatment option. While patients show high response rate to standard chemotherapy such as cisplatin/etoposide, they soon develop drug resistance and disease progression. Therefore, new therapeutic strategies are urgently needed for SCLC. BPR6K609S0 is a novel Aurora kinase inhibitor which has been designed to inhibit the kinase activity of Aurora A, and induces proteasome-mediated degradation of MYC. The BPR6K609S0 active molecule provokes cell apoptosis and inhibits proliferation of several SCLC cell lines with IC50 & lt; 100 nM. Oral administration of BPR6K609 induces & gt;60 % tumor regression in a NCI-H446 xenograft mouse model. In addition, BPR6K609 further reduces tumor progression in NCI-H446 xenograft mice pre-treated with LY3295668, an Aurora A-selective inhibitor which is currently under clinical investigation. These results support the clinical potential of BRP6K609S0 for the treatment of SCLC. Citation Format: Ya-Hui Chi, Chun-Ping Chang, Yi-Yu Ke, Wen-Hsing Lin, Wan-Ping Wang, Chia-Hua Tsai, Yen-Ting Chen, Yu-Jie Su, Ming-Chun Hung, Zhong-Wei Wu, Mine-Hsine Wu, Teng-Kuang Yeh, Ching-Ping Chen, Jen-Shin Song, Chiung-Tong Chen, Chuan Shih. BPR6K609: An Aurora kinase inhibitor targeting small cell lung cancer with MYC amplification [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 1937.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 4413-4413
    Abstract: Drug delivered by conjugate with antibody is expected to increase drug concentrations at the tumor sites for improved antitumor effects. Showing some successes, the antibody-drug conjugate system has certain limitations such as difficulties in protein characterization, high cost and individual variations in expression of the epitope, which might abrogate the antibody recognition and cause premature drug release arising from antibody-drug linker instability and thus ineffectiveness. Phosphatidylserine (PS) exists in bulk amount in the tumor microenvironment. Studies showed that a small sized Zinc (II)-dipicolylamine (Zn-DPA) molecule was superior in locating PS surrounding the apoptotic sites in vivo. Although Zn-DPA has been constructed incorporating fluorescent dyes as optical tools for imaging the PS-exposed cell membranes to identify tumor sites from healthy cells in animals, drug conjugates with Zn-DPA has never been described. We aimed to design, synthesize and evaluate a Zinc-dipicolylAmine directed Pharmaceutical delivery System (ZAPS), a platform for spatial- and temporal-release of drug specifically at the targeted tumor site. Zn-DPA was conjugated through standard coupling conditions with various linkers to SN-38 and the Zn-DPA-linker-SN-38 conjugates were obtained and investigated for structure-activity relationships on the chemical stability in plasma and cytotoxicity against cancer cells. Promising ZAPS-SN-38 conjugates were evaluated for activities against tumor growths in nude mice. CPT-11 and SN-38 were included for comparisons. ZAPS-SN-38 conjugates were examined for tolerable doses in mice. Among the ZAPS-SN-38 conjugates synthesized, ZAPS001 was found chemically stable in mouse plasma and in vitro active against several cancer cells. ZAPS001 dose-dependently inhibited the growth of Colo205 tumors in nude mice. Zn-DPA-linker001 (ZAPS001 without SN-38) showed no inhibition effect on the tumor growth. Intriguingly, employing only 40% of the SN-38 delivered by CPT-11 (40 mg/kg), ZAPS001 resulted in 8-fold increase in antitumor activity compared to that of CPT-11 given at the same dose regimen. Furthermore, ZAPS001 is also active against pancreatic Mia-Paca2 and BxPC-3 tumors in mice. An increased SN-38 level delivered by ZAPS001 to the tumors in mice was also observed. ZAPS is thus capable of selectively associating with PS-exposing tumor cells/tissues to achieve site-specific delivery of anticancer agents. Our findings strongly support that ZAPS conferred an “in situ dose amplification” effect, i.e., the cytotoxics-induced PS exposure could specifically recruit more ZAPS-drug conjugates to the tumor site and further enhance its therapeutic effect. ZAPS provides spatial and temporal controls to increase drug concentration at targeted disease sites, reduce drug dosage, lessen the toxic side effects, and thus increase therapeutic index of the anticancer drugs. Citation Format: Lun K. Tsou, Yu-Wei Liu, Yun-Yu Chen, Chen-Fu Lo, Teng-Kuang Yeh, Chien-Huang Wu, Kak-Shan Shia, Joe C. Shih, Brian D. Gary, Koon Y. Pak, Chiung-Tong Chen. Zinc-dipicolylamine directed pharmaceutical delivery system (ZAPS) as an innovative cancer drug delivery platform. [abstract] . In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4413. doi:10.1158/1538-7445.AM2015-4413
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 3851-3851
    Abstract: Small cell lung cancer (SCLC) is one the most aggressive tumors with poor survival rate. SCLC patients often present with metastasis at time of diagnosis, excluding surgery as a treatment option. While patients show high response rate to standard chemotherapy such as cisplatin/etoposide, they soon develop drug resistance and disease progression. Therefore, new therapeutic strategies are urgently needed for SCLC. Several characteristics of SCLC such as aggressiveness and drug resistance could attribute to the existence of a cancer stem cell (CSC) subpopulation in SCLC. The SCLC cell line NCI-H446 has been shown to present high degree of stemness and express stem cell markers including CD133, OCT4, MYC and Nestin. Here we develop a pyrimidine-based small molecule BPR6K471 which potently inhibits Aurora kinase activities in enzymatic- and cell- based assays. BPR6K471 efficiently (IC50 = 66 nM) inhibits proliferation of NCI-H446, and reduces the expression of stem-cell markers. In addition, intravenous injection of BPR6K471 inhibits & gt;90 % progression of NCI-H446 in a mouse xenograft model. These results suggest that targeting Aurora kinases may be a potential therapeutic strategy to combat the CSC subpopulation in SCLC. Citation Format: Chun-Ping Chang, Yi-Yu Ke, Wen-Hsing Lin, Dai-Hui Jhuo, Wan-Ping Wang, Chia-Hua Tsai, Yen-Ting Chen, Yu-Jie Su, Ming-Chun Hung, Zhong-Wei Wu, Po-Chu Kuo, Teng-Kuang Yeh, Ching-Ping Chen, Jen-Shin Song, Chiun-Tong Chen, Chuan Shih, Ya-Hui Chi. An Aurora kinase inhibitor BPR6K471 inhibits tumor growth and reduces the cancer stem cell-like properties of small cell lung 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 3851.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1756-1756
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1756-1756
    Abstract: Lung cancer is the leading cause of cancer death in the world and non-small cell lung cancer (NSCLC) accounts for 85% of the lung cancer deaths. In recent years, EGFR tyrosine kinase inhibitors (TKIs) have showed remarkable effects in patients with certain genetic alternations in the EGFR proteins. However, the clinical benefits of two first-generation EGFR TKIs (gefitinib and erlotinib) have been limited due to the acquired resistance from patients within 9-12 months after treatment, therefore, the discovery of next-generation EGFR-TKIs poses an utmost priority. Our team at IBPR has identified DBPR112 as a potent EGFR-TKI, showing IC50 of 2 nM in HCC827 cells, EGFRWild-Type (IC50: 10 nM) and EGFRL858R/T790M (IC50: 70 nM), which are comparatively better than gefitinib and similar to that of BIBW2992 (afatinib, developed by Boehringer Ingelheim, and approved by FDA in 2013). DBPR112 (10, 50 and 100 mg/kg/day, 5 days/wk for 2 weeks) was orally effective against the growth of human lung HCC827 tumors subcutaneously xenografted in nude mice. A dramatic reduction in tumor size was noted with DBPR112 treatment at 50 and 100 mg/kg/day, while displaying negligible body weight loss in all dosing groups. In addition, the pharmacokinetics properties of DBPR112 are superior to those of BIBW2992; demonstrating the potential of DBPR112 as a therapeutic candidate for the treatment of lung adenocarcinoma with EGFR mutations. This NRPB-granted 4-year top-down project was commenced in Dec. 2012 and aimed at accomplishing a comprehensive program of pre-clinical development and phase I study. To date, several pre-clinical studies and progress were steadily accomplished, including non-GMP production of 5 kg DBPR112 (tox-lot), preliminary analysis methods development, pre-formulation study, pharmacokinetics/metabolism studies and preliminary toxicology evaluation. GLP-toxicity studies, GMP-production of DBPR112 (clinical-lot) and formulation for clinical use will be scheduled in 2014 (2nd year of this top-down project), and we look forward to filing an IND application in early 2015. Citation Format: Hui-Yi Shiao, Tsu-An Hsu, Wen-Hsing Lin, Tsong-Toh Yang, Hui-Fang Hsieh, Chiung-Tong Chen, Teng-Kuang Yeh, Hsing-Pang Hsieh. Novel irreversible EGFR tyrosine kinase inhibitor, DBPR112, as a therapeutic candidate for lung adenocarcinoma. [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 1756. doi:10.1158/1538-7445.AM2014-1756
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 523-523
    Abstract: Upregulation of Aurora kinases has been associated with increased tumor progression, and thus they are appealing targets for the development of anti-cancer therapies. In addition to have a critical role in cell cycle regulation, Aurora kinases have been shown to stabilize MYC-family oncoproteins for the maintenance of malignancy. Aurora kinase inhibitors such as Alisertib has demonstrated compelling anti-tumor efficacies; however, the reported side effects including serious haematological disturbances have limited its risk-benefit ratio in clinical use. In this study we discovered a novel pyrimidine-based Aurora kinase inhibitor compound A that degraded MYC- and MYCN- oncoproteins with better potency than Alisertib. The acyl-based prodrug design leads to the discovery of compound B which was able to regress MYC- or MYCN- overexpressing tumor xenografts including small cell lung cancer, neuroblastoma, hepatocellular carcinoma and medulloblastoma using a once-a-week (QW) oral dosing regimen. Pharmacokinetic studies revealed that the tumor/plasma ratio of compound B was about 20 at 24 h post drug administration, and the active compound remained detectable in the tumors after 7 days. No significant haematological or liver/kidney biochemical aberration was observed in mice treated with up to 500 mg/kg of DBPR728 on a QW dosing regimen in a 21-day cycle. The unique pharmacokinetic and molecular properties of compound B hold potential clinical promise for treating MYC- and MYCN- amplified tumors with manageable on-target haematological adverse effects caused by Aurora kinase inhibition. Citation Format: Ya-Hui Chi, Chun-Ping Chang, Teng-Kuang Yeh, Wan-Ping Wang, Yen-Ting Chen, Chia-Hua Tsai, Yan-Fu Chen, Yi-Yu Ke, Jing-Ya Wang, Ching-Ping Chen, Tsung-Chih Hsieh, Mine-Hsine Wu, Chiung-Tong Chen. Targeting myc-amplified cancers with a novel prodrug inhibiting aurora A kinase [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 523.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 227-227
    Abstract: Zinc(II)-dipicolylamine (Zn-DPA) have been described to specifically complex with phosphatidylserine (PS), which has a higher level at the external surface of cancer cells in tumorigenic condition. BPRDP056 is a Zn-DPA-SN38 conjugate designed to provide a PS-targeting drug delivery of cytotoxic payload SN-38 at the tumor microenvironment, thereby, decrease the dosage of SN38, while induce apoptosis in cancer cells. The in vivo therapeutic efficacy of BPRDP056 against the growths of human tumors has been shown significant in mice subcutaneously bearing a tumor type of pancreas, prostate, colon, liver, breast and glioblastoma, as well as in mice with an orthotopic pancreatic tumor. BPRDP056 shrunk tumors at a lower dosing intensity (~20%) of SN38 compared to CPT-11 in all models tested. Micro-Western assays showed that BPRDP056 exhibited apoptotic cell death signal levels similar to those of CPT-11 in the treated tumors in mice. Furthermore, pharmacokinetic and preliminary toxicology studies showed that BPRDP056 has a good stability in circulation with an acceptable therapeutic safety window in mice. BPRDP056 has been demonstrated with a tumor targeting ability and thus increases the cytotoxic payload SN38 concentration in situ for improved efficacy. Its therapeutic spectrum against malignant neoplasm will be expected to cover the PS-rich tumor microenvironment of all cancer types. BPRDP056 is a first-in-class Small Molecule Drug Conjugate for anti-cancer therapy. Citation Format: Chia-Yu Hsu, Yun-Yu Chen, Chen-Fu Lo, Tai-Yu Chiu, Ching-Ping Chen, Chen-Lung Huang, Chung-Yu Huang, Min-Hsien Wang, Yu-Sheng Chao, Joe C. Shih, Teng-Kuang Yeh, Lun K. Tsou, Chiung-Tong Chen. BPRDP056, a novel small molecule drug conjugate specifically targeting phosphatidylserine for cancer therapy [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 227.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2004
    In:  Clinical Cancer Research Vol. 10, No. 22 ( 2004-11-15), p. 7677-7684
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 10, No. 22 ( 2004-11-15), p. 7677-7684
    Abstract: Purpose: The present report describes the development of paclitaxel-loaded gelatin nanoparticles for use in intravesical therapy of superficial bladder cancer. The commercial formulation of paclitaxel contains Cremophor, which forms micelles and thereby entraps the drug and reduces its partition across the urothelium. Experimental Design: Paclitaxel-loaded gelatin nanoparticles were prepared using the desolvation method, and their physicochemical and biological properties were characterized. Results: The size of the particles ranged from 600 to 1,000 nm and increased with the molecular weight of the gelatin polymer. Under optimal conditions, the yield was & gt;80%, and the drug loading was 0.7%. Wide-angle X-ray diffraction analysis showed that the entrapped paclitaxel was present in an amorphous state, which has higher water solubility compared with the crystalline state. Identical, rapid drug release from nanoparticles was observed in PBS and urine, with ∼90% released at 37°C after 2 hours. Treatment with a protease (i.e., Pronase) rapidly degraded the nanoparticles, with half-lives of 23.8 minutes, 0.6 minute, and 0.4 minute in the presence of 0.01, 0.05, and 0.25 mg/mL Pronase, respectively. The paclitaxel-loaded nanoparticles were active against human RT4 bladder transitional cancer cells; the IC50 paclitaxel-equivalent concentrations were nearly identical to those of aqueous solutions of paclitaxel, i.e., ∼30 nmol/L (equivalent to ∼25 ng/mL) for 2-hour treatments and ∼4 nmol/L for 96-hour treatments. In dogs given an intravesical dose of paclitaxel-loaded particles, the drug concentrations in the urothelium and lamina propria tissue layers, where Ta and T1 tumors would be located, were 7.4 ± 4.3 μg/g (mean ± SD; 3 dogs; 9 tissue sections), which were 2.6× the concentrations we reported for dogs treated with the Cremophor formulation. Conclusions: Paclitaxel-loaded gelatin nanoparticles represent a rapid release, biologically active paclitaxel formulation that can be used for intravesical bladder cancer therapy.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2004
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1664-1664
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1664-1664
    Abstract: Indoleamine 2,3-dioxygenase has been considered as a promising anti-cancer drug target. Several pharmaceutical companies, including Pfizer, Merck, and Bristol-Myers Squibb, are known to be in pursuit of IDO inhibitors, and Incyte recently reported good results in the phase II clinical trial of the IDO inhibitor Epacadostat. In previous work, we developed a series of IDO inhibitors based on a high-throughput screening core structure, sulfonylhydrazide. Further, we developed the 4-bromophenylhydrazinyl benzenesulfonylphenylurea, as a potent IDO inhibitor with an IC50 value within 100 nM in inhibiting IDO in vitro. This compound demonstrated a 30% reduction in tumor weight in a murine CT26 syngeneic model on day 18 with 100 mg/kg oral administration twice daily, indicating that the 4-bromophenylhydrazinyl benzenesulfonylphenylurea may have potential for further investigation in the development of anti-tumor drugs. Citation Format: Shau-Hua Ueng, Ching-Chuan Kuo, Shu-Yu Lin, Teng-Kuang Yeh, Jen-Shin Song, Ming-Shiu Hung, Chiung-Tong Chen. 4-Bromophenylhydrazinyl benzenesulfonylphenylureas inhibit indoleamine 2,3-dioxygenase in vitro and in vivo [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 1664.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages