• Title/Summary/Keyword: paclitaxel '

Search Result 331, Processing Time 0.026 seconds

Clinical Comparison between Paclitaxel Liposome (Lipusu®) and Paclitaxel for Treatment of Patients with Metastatic Gastric Cancer

  • Xu, Xu;Wang, Lin;Xu, Huan-Qin;Huang, Xin-En;Qian, Ya-Dong;Xiang, Jin
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.14 no.4
    • /
    • pp.2591-2594
    • /
    • 2013
  • Aim: To compare the efficacy and safety of paclitaxel liposome (Lipusu$^{(R)}$) with paclitaxel in combination with tegafur and oxaliplatin in treating patients with advanced gastric cancer. Materials and Methods: Patients with advanced gastric cancer receiving chemotherapy were retrospectively collected, and divided into two groups. Patients in group A received paclitaxel liposomes at a dose of 135 $mg/m^2$ on day 1 of each cycle, and patients in group B were given paclitaxel at the same dose with the same timing. All patients received tegafur at a dose of 500 $mg/m^2$ on days 1-5, and oxaliplatin at a dose of 80-100 $mg/m^2$ on day 1 for 2 cycles (each cycle was 21 d in total). Results: Fifty-eight patients could be evaluated for efficacy. The overall response rate was 47% in group A (14/30), and 46% in group B (13/28). Disease control rate was 73% in group A (22/30), and 71% in group B (20/28) (P>0.05). No significant differences were detected in hematologic and neurologic toxicities between the two groups (P>0.05). However, nausea, vomiting and hypersensitive reactions were significantly lower in group A than in group B (P<0.05). Conclusion: Paclitaxel liposomes are as effective as paclitaxel when combined with tegafur and oxaliplation in treating patients with advanced gastric cancer, but adverse reactions with paclitaxel liposomes are less common.

Pharmacophore Models of Paclitaxel- and Epothilone-Based Microtubule Stabilizing Agents

  • Lee, Sangbae;Lee, Yuno;Briggs, James M.;Lee, Keun Woo
    • Bulletin of the Korean Chemical Society
    • /
    • v.34 no.7
    • /
    • pp.1972-1984
    • /
    • 2013
  • Microtubules play an important role in intracellular transport, mobility, and particularly mitosis. Paclitaxel (Taxol$^{TM}$) and paclitaxel-like compounds have been shown to be anti-tumor agents useful for various human tumors. Paclitaxel-like compounds operate by stabilizing microtubules through interface binding at the interface between two ${\beta}$-tubulin monomers in adjacent protofilaments. In this paper we present the elucidation of the structural features of paclitaxel and paclitaxel-like compounds (e.g., epothilones) with microtubule stabilizing activities, and relate their activities to spatial and chemical features of the molecules. CATALYST program was used to generate three-dimensional quantitative structure activity relationships (3D-QSARs) resulting in 3D pharmacophore models of epothilone- and paclitaxel-derivatives. Pharmacophore models were generated from diverse conformers of these compounds resulting in a high correlation between experimental and predicted biological activities (r = 0.83 and 0.91 for epothilone and paclitaxel derivatives, respectively). On the basis of biological activities of the training sets, five- and four-feature pharmacophore hypotheses were generated in the epothilone and paclitaxel series. The validation of generated hypotheses was achieved by using twelve epothilones and ten paclitaxels, respectively, which are not in the training sets. The clustering (grouping) and merging techniques were used in order to supplement spatial restrictions of each of hypothesis and to develop more comprehensive models. This approach may be of use in developing novel inhibitor candidates as well as contributing a better understanding of structural characters of many compounds useful as anticancer agents targeting microtubules.

Comparison of the Neointima Inhibition Between Paclitaxel- and Sirolimus-Eluting Expanded Polytetrafluoroethylene Hemodialysis Grafts in a Porcine Model

  • Baek, Insu;Cho, AJin;Hwang, Jinsun;Kim, Heasun;Park, Jong-Sang;Kim, Dae Joong
    • Bulletin of the Korean Chemical Society
    • /
    • v.34 no.6
    • /
    • pp.1663-1667
    • /
    • 2013
  • Neointimal hyperplasia causes vascular access dysfunction in hemodialysis patients with synthetic arteriovenous (AV) grafts. Several studies have reported that paclitaxel- or sirolimus-eluting AV grafts inhibit neointimal hyperplasia and display lower rates of stenosis compared with control grafts. However, there have been few comparative studies of the efficacy of paclitaxel- and sirolimus-eluting grafts. We compared the neointimal hyperplasia of paclitaxel- and sirolimus-eluting grafts. AV grafts were implanted laterally between the common carotid artery and the external jugular vein in 12 female Landrace pigs. The animals were sacrificed six weeks after surgery. The neointimal hyperplasia at the anastomosis sites of the grafts was quantified using the ratio of the intragraft hyperplasia to the graft area (H/G ratio) at the graft-vessel interface. The area of intimal hyperplasia at the venous (paclitaxel 1.06 [0.72-1.56] vs sirolimus 2.40 [1.72-3.0] $mm^2$, P = 0.04) and arterial anastomosis sites (paclitaxel 0.93 [0.57-1.48] vs sirolimus 2.40 [1.72-3.0] $mm^2$, P = 0.04) was significantly different between the two groups. However, the H/G ratios for the venous anastomosis site (paclitaxel 0.25 (0.17-0.38) vs sirolimus 0.38 (0.2-0.66), P = 0.4) and the arterial anastomosis site (paclitaxel 0.19 (0.08-0.39) vs sirolimus 0.41 (0.34-0.50), P = 0.1) did not differ significantly between the groups. In conclusion, there was no significant difference in the inhibition of neointimal hyperplasia by sirolimus- and paclitaxel-eluting AV grafts.

Sensitization of Cervical Carcinoma Cells to Paclitaxel by an IPP5 Active Mutant

  • Zeng, Qi-Yan;Huang, Yu;Zeng, Lin-Jie;Huang, Min;Huang, Yong-Qi;Zhu, Qi-Fang
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.19
    • /
    • pp.8337-8343
    • /
    • 2014
  • Paclitaxel is one of the best anticancer agents that has been isolated from plants, but its major disadvantage is its dose-limiting toxicity. In this study, we obtained evidence that the active mutant IPP5 ($8-60hIPP5^m$), the latest member of the inhibitory molecules for protein phosphatase 1, sensitizes human cervix carcinoma cells HeLa more efficiently to the therapeutic effects of paclitaxel. The combination of $8-60hIPP5^m$ with paclitaxel augmented anticancer effects as compared to paclitaxel alone as evidenced by reduced DNA synthesis and increased cytotoxicity in HeLa cells. Furthermore, our results revealed that $8-60hIPP5^m$ enhances paclitaxel-induced G2/M arrest and apoptosis, and augments paclitaxel-induced activation of caspases and release of cytochrome C. Evaluation of signaling pathways indicated that this synergism was in part related to downregulation of NF-${\kappa}B$ activation and serine/threonine kinase Akt pathways. We noted that $8-60hIPP5^m$ downregulated the paclitaxel-induced NF-${\kappa}B$ activation, $I{\kappa}B{\alpha}$ degradation, PI3-K activity and phosphorylation of the serine/threonine kinase Akt, a survival signal which in many instances is regulated by NF-${\kappa}B$. Together, our observations indicate that paclitaxel in combination with $8-60hIPP5^m$ may provide a therapeutic advantage for the treatment of human cervical carcinoma.

Injection Formulation of Paclitaxel Employing Solid Lipid Nanoparticles (SLN) (고형지질나노입자를 이용한 파클리탁셀의 주사제 설계)

  • Choi, Sung-Up;Kim, Sun-Kyu;Lee, Jung-Min;Choi, Young-Wook
    • Journal of Pharmaceutical Investigation
    • /
    • v.33 no.4
    • /
    • pp.319-322
    • /
    • 2003
  • Many studies have been attempted to overcome the problems of paclitaxel related to the extremely low aqueous solubility of paclitaxel and the unexpected side-effects caused by $Cremophor^{\circledR}$ EL in a commercial paclitaxel formulation, $Taxol^{\circledR}$. In order to formulate a new delivery system suitable for intravenous administration without toxic excipients, in this study, paclitaxel was incorporated into solid lipid nanoparticles (Px-SLN) by hot homogenization technique using a microfluidizer. Particle size and zeta potential were measured by a Zetasizer. In vitro drug release experiment was performed by a dialysis diffusion method. Each Px-SLN or $Taxol^{\circledR}$ was intravenously administered to the male Sprague-Dawley rats at a dose of 5 mg/kg as paclitaxel. Blood samples were deproteinated with acetonitrile and assayed for paclitaxel by the validated HPLC/MS/MS method. Mean particle size and zeta potential were measured as 72.1 nm (< Polydispersity 0.3) and -41.5 mV, respectively. The content of paclitaxel in SLN was 1.42 mg/ml and the drug loading efficiency was $71.2{\pm}4.3%$. The $AUC_t$ of Px-SLN was 3.4-fold greater than that of $Taxol^{\circledR}$. The Px-SLN might be a promising candidate for an alternative formulation for the parenteral delivery of paclitaxel.

Pharmacokinetic Interaction between Diltiazem and Paclitaxel in Rats (딜티아젬과 파크리탁셀의 약물동태학적 상호작용)

  • Choi, Jun-Shik;Kim, Hyung-Joong
    • Journal of Pharmaceutical Investigation
    • /
    • v.33 no.4
    • /
    • pp.299-304
    • /
    • 2003
  • The purpose of this study was to investigate the effect of coadministration (2.5, 10, 20 mg/kg) and 3 or 7 days-pretreatment (10 mg/kg) of diltiazem on the pharmacokinetic parameters of paclitaxel (50 mg/kg) given orally in rats. The plasma concentrations of paclitaxel coadministered or pretreated with diltiazem were significantly (p<0.05 at 20 mg/kg coadmin., p<0.05 at pretreat.) increased compared to that of control, from 0.5 hr to 24 hr. Area under the plasma concentration-time curve (AUC) of paclitaxel coadministered or pretreated with diltiazem was significantly (p<0.05 at 20 mg/kg coadmin., p<0.01 at pretreat.) higher than that of control. Peak concentrations $(C_{max})$ of paclitaxel with diltiazem were significantly (p<0.05 at 20 mg/kg coadmn. and pretreat.) increased compared to that of control. Elimination rate constants $(K_{el})$ of paclitaxel with diltiazem were significantly (p<0.05 at 20 mg/kg and 7 days-pretreat.) reduced compared to those of control. Half-life $(t_{1/2})$ and mean residence time (MRT) of paclitaxel with diltiazem was significantly (p<0.05 at 20 mg/kg ad 7 days-pretreat.) prolonged compared to those of control. Absolute bioavailability (AB%) of paclitaxel with diltiazem was significantly (p<0.05 at 20 mg/kg and 3 days-pretreat, p<0.01 at 7 days -pretreat.) increased compared to that of control. Based on these results, it might be considered that diltiazem may inhibit cytochrome $P_{450}$ and P-glycoprotein, which are respectively engaged in paclitaxel absorption and metabolism in liver and gastrointestinal mucosa.

ATG5 knockout promotes paclitaxel sensitivity in drug-resistant cells via induction of necrotic cell death

  • Hwang, Sung-Hee;Yeom, Hojin;Lee, Michael
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.24 no.3
    • /
    • pp.233-240
    • /
    • 2020
  • Autophagy regulators are often effective as potential cancer therapeutic agents. Here, we investigated paclitaxel sensitivity in cells with knockout (KO) of ATG5 gene. The ATG5 KO in multidrug resistant v-Ha-ras-transformed NIH 3T3 cells (Ras-NIH 3T3/Mdr) was generated using the CRISPR/Cas9 technology. The qPCR and LC3 immunoblot confirmed knockout of the gene and protein of ATG5, respectively. The ATG5 KO restored the sensitivity of Ras-NIH 3T3/Mdr cells to paclitaxel. Interestingly, ATG5 overexpression restored autophagy function in ATG5 KO cells, but failed to rescue paclitaxel resistance. These results raise the possibility that low level of resistance to paclitaxel in ATG5 KO cells may be related to other roles of ATG5 independent of its function in autophagy. The ATG5 KO significantly induced a G2/M arrest in cell cycle progression. Additionally, ATG5 KO caused necrosis of a high proportion of cells after paclitaxel treatment. These data suggest that the difference in sensitivity to paclitaxel between ATG5 KO and their parental MDR cells may result from the disparity in the proportions of necrotic cells in both populations. Thus, our results demonstrate that the ATG5 KO in paclitaxel resistant cells leads to a marked G2/M arrest and sensitizes cells to paclitaxel-induced necrosis.

Development of High Performance Liquid Chromatography for Paclitaxel Purification from Plant Cell Cultures

  • Kim, Jin-Hyun;Choi, Hyung-Kyoon;Hong, Seung-Suh;Lee, Hyun-Soo
    • Journal of Microbiology and Biotechnology
    • /
    • v.11 no.2
    • /
    • pp.204-210
    • /
    • 2001
  • Paclitaxel can be produced in high yield and with a high degree of purify from plant cell cultures of Taxus chinensis. The complete purification method was systematically established and described. This method was an efficient procedure for the purification of paclitaxel from crude paclitaxel, consisting or reverse-phase chromatography, followed by a normal-phase chromatography. The two-stage HPLC purification scheme serves as an effective and economical approach for resolving paclitaxel from complex mixtures of taxoids, with high purify (>99%) and low impurities (<0.1%). The process is readily scalable to a pilot plant and eventually to a production environment where multikilogram quantities of material are expected to be produced. The process has been optimized to minimize solvent usage, complexity, and operating costs.

  • PDF

Anti-tumor Activity of Paclitaxel Prodrug Conjugated with Polyethylene Glycol

  • Lee, Keyong-Ho;Chung, Yong-Jun;Kim, Youn-Chul;Song, Seog-Jeong
    • Bulletin of the Korean Chemical Society
    • /
    • v.26 no.7
    • /
    • pp.1079-1082
    • /
    • 2005
  • The purpose of this study was to develop a method for increasing the solubility of paclitaxel in water to reduce its toxicity and make the drug more feasible for chemotherapy. A series of highly water soluble paclitaxel polyethylene glycol (PEG) esters were synthesized and evaluated for their anti-tumor activity and toxicity. The solubility of 7-polyethylene glycol paclitaxel carbonate 5 was 840 mg/mL and the acute toxicity ($LD_{50}$) was 286 mg/kg. Because of its reduced toxicity, compound 5 showed a dramatic reduction of tumor volume without any loss of animals in long-term treatment (daily consecutive injections for 15 days).

In Vitro Study of Tumor Seeking Radiopharmaceutical Uptake by Human Breast Cancer Cell Line MCF-7 after Paclitaxel Treatment (사람 유방암세포주 MCF-7에 Paclitaxel 처치 후 종양영상용 방사성의약품 섭취 변화에 대한 시험관내 연구)

  • Choi, Joon-Young;Choi, Yong;Choe, Yearn-Seong;Lee, Kyung-Han;Kim, Byung-Tae
    • Nuclear Medicine and Molecular Imaging
    • /
    • v.41 no.5
    • /
    • pp.364-372
    • /
    • 2007
  • Purpose: This study was designed to investigate the cellular uptake of various tumor imaging radiopharmaceuticals in human breast cancer cells before and after paclitaxel exposure considering viable cell number. Materials and Methods: F-18-fluorodeoxyglucose, C-11-methionine, Tl-201, Tc-99m-MIBI, and Tc-99m-tetrofosmin were used to evaluate the cellular uptake in MCF-7 cells. MCF-7 cells were cultured in multi-well plates. Wells were divided into DMSO exposure control group, and paclitaxel exposure group. The exposure durations of paclitaxel with 10 nM or 100 nM were 2 h, 6 h, 12 h, 24 h, and 48 h. Results: Viable cell fraction was reduced as the concentration and exposure time of paclitaxel increased. After 10 nM paclitaxel exposure, the cellular uptake of all 5 radiopharmaceuticals was not reduced significantly, irrespective of exposure time and viable cell fraction. After 100 nM paclitaxel exposure, the cellular uptake of all 5 radiopharmaceuticals was enhanced significantly irrespective of viable cell fraction. The peak uptake was observed in experimental groups with paclitaxel exposure for 6 to 48 h according the type of radiopharmaceutical. When the cellular uptake was adjusted for the viable cell fraction and cell count, the peak cellular uptake was observed in experimental groups with paclitaxel exposure for 48 h, irrespective of the type of radiopharmaceutical. Conclusion: The cellular uptake of F-18-fluorodeoxyglucose, C-11-methionine, Tl-201, Tc-99m-MIBI, and Tc-99m-tetrofosmin did not reflect viable cell number in MCF-7 cells after paclitaxel exposure for up to 48 h.