Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Chem Commun (Camb) ; 59(61): 9352-9355, 2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37431730

ABSTRACT

Transarterial chemoembolization (TACE) is one of the most commonly used treatments for hepatocellular carcinoma (HCC); however, the poor stability of emulsified chemotherapy drugs by iodinated oil always leads to serious systemic cytotoxicity. Herein, a composite hydrogel Epi/Etpoil@MC/XG was proposed by stably distributing ethiodized poppyseed oil (Etpoil) and epirubicin (Epi) in the blend hydrogel of methylcellulose (MC) and xanthan gum (XG). Benefiting from its adjusted thermo-responsive and injectable properties, the Epi/Etpoil@MC/XG has been successfully applied in the embolization of the feeding artery for a VX2 tumor model.


Subject(s)
Carcinoma, Hepatocellular , Chemoembolization, Therapeutic , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Hydrogels/therapeutic use , Epirubicin/pharmacology , Epirubicin/therapeutic use , Ethiodized Oil/therapeutic use , Arteries
2.
J Mater Chem B ; 11(2): 415-429, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36512437

ABSTRACT

Ferroptosis, a type of programmed cell death induced by the iron-dependent lipid hydroperoxide pathway, has attracted widespread attention. However, Fenton response-dependent ferroptosis has many limitations, such as insufficient reaction conditions in the tumor micro-environment. Here, we propose an all-in-one phototherapy nanoplatform consisting of iron-polydopamine (Fe-PDA), a folic acid-modified red blood cell membrane (FA-RBCm), and epirubicin (EPI), namely, Fe-PDA-EPI@FA-RBCm NPs, to achieve enhanced photothermal-ferroptosis effects via overcoming the limitations of the Fenton-like reaction. The results showed that the synthesized biomimetic nanoparticles could decompose hydrogen peroxide (H2O2) to generate hydroxyl radicals (˙OH), and further induce the non-apoptotic ferroptosis pathway. After irradiation with near-infrared (NIR) light, the uptake of Fe-PDA-EPI@FA-RBCm NPs by cells could be effectively promoted, and it presented impressive in vitro and in vivo photothermal properties. In vitro and in vivo results showed that laser irradiation could enhance ferroptosis by promoting the production of reactive oxygen species (ROS) and lipid peroxides, down-regulating the expression of glutathione peroxidase 4 (GPX4), and reducing the mitochondrial membrane potential. Furthermore, the photothermal-promoted ferroptosis and apoptosis pathways (photothermal therapy and chemotherapy) exhibited outstanding synergistic antitumor efficacy in vitro and in vivo, with an in vivo tumor inhibition rate as high as 76.95%. In conclusion, the construction of tumor-targeted biomimetic nanocarriers utilizing the advantageous properties of RBCm has been investigated as a potential anticancer strategy.


Subject(s)
Ferroptosis , Nanoparticles , Neoplasms , Hydrogen Peroxide/pharmacology , Nanoparticles/therapeutic use , Apoptosis , Epirubicin/pharmacology , Iron/pharmacology
3.
Folia Med (Plovdiv) ; 63(4): 488-495, 2021 Aug 31.
Article in English | MEDLINE | ID: mdl-35851170

ABSTRACT

AIM: We evaluated the tumor-inhibiting effect of artemisinin applied separately and in combination with epirubicin on leukemia HL-60 and HL-60/Dox cell lines, its dose modulation effect and its potency to  influence iron-induced oxidative damage of biologically relevant molecules. MATERIALS AND METHODS: MTT assay and the method of Chou-Talalay were used to show the inhibition of tumor cell proliferation and to evaluate the synergistic effect and modulation effect of artemisinin and epirubicin at varying concentrations. We also used spectrophotometric assays to determine the potency of artemisinin to influence iron-induced molecular degradation of lecithin and deoxyribose. RESULTS: Artemisinin exhibits tumor-inhibiting effect on both the anthracycline-sensitive and anthracycline-resistant promyelocytic cell lines, reaching 88% and 61% (T/C), respectively, when applied at higher concentrations in a dose-dependent manner. The combination of artemisinin and epirubicin shows synergistic effects in all tested concentrations on doxorubicin-resistant cells (CI<0.7). Artemisinin sensitizes the resistant cells towards epirubicin as shown by the CI (combination index) values and has a dose-modulation effect as shown by DRI (dose reduction index). Artemisinin induces deoxyribose oxidative degradation when applied alone and exerts synergistic deoxyribose degradation effect when applied with iron. However, artemisinin does not influence the studied processes in the lecithin-containing model system and has no potential to induce lipid peroxidation. CONCLUSIONS: This study presents a new opportunity to enhance the effectiveness of epirubicin-based treatment regimens with addition of artemisinins for resistant tumors.


Subject(s)
Artemisinins , Leukemia , Anthracyclines , Artemisinins/pharmacology , Deoxyribose , Doxorubicin/pharmacology , Drug Synergism , Epirubicin/pharmacology , Humans , Iron , Lecithins , Leukemia/drug therapy
4.
Int J Cancer ; 147(9): 2515-2525, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32488909

ABSTRACT

Antiangiogenic drugs are potentially a useful supplement to neoadjuvant chemotherapy for a subgroup of patients with human epidermal growth factor receptor 2 (HER2) negative breast cancer, but reliable biomarkers for improved response are lacking. Here, we report on a randomized phase II clinical trial to study the added effect of bevacizumab in neoadjuvant chemotherapy with FEC100 (5-fluorouracil, epirubicin and cyclophosphamide) and taxanes (n = 132 patients). Gene expression from the tumors was obtained before neoadjuvant treatment, and treatment response was evaluated by residual cancer burden (RCB) at time of surgery. Bevacizumab increased the proportion of complete responders (RCB class 0) from 5% to 20% among patients with estrogen receptor (ER) positive tumors (P = .02). Treatment with bevacizumab was associated with improved 8-year disease-free survival (P = .03) among the good responders (RCB class 0 or I). Patients treated with paclitaxel (n = 45) responded better than those treated with docetaxel (n = 21; P = .03). Improved treatment response was associated with higher proliferation rate and an immune phenotype characterized by high presence of classically activated M1 macrophages, activated NK cells and memory activated CD4 T cells. Treatment with bevacizumab increased the number of adverse events, including hemorrhage, hypertension, infection and febrile neutropenia, but despite this, the ECOG status was not affected.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bevacizumab/pharmacology , Breast Neoplasms/therapy , Neoadjuvant Therapy/methods , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Breast/cytology , Breast/pathology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Chemotherapy, Adjuvant/methods , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Disease-Free Survival , Epirubicin/pharmacology , Epirubicin/therapeutic use , Female , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Follow-Up Studies , Humans , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/immunology , Mastectomy , Middle Aged , Neoplasm, Residual , Norway/epidemiology , Receptor, ErbB-2/analysis , Receptor, ErbB-2/metabolism , Tumor Burden/drug effects , Tumor Burden/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
5.
Anticancer Res ; 40(2): 915-921, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32014935

ABSTRACT

BACKGROUND/AIM: This study aimed was to clarify the impact of pegfilgrastim (PEG) 3.6 mg primary prophylaxis of febrile neutropenia (FN) on the average relative dose intensity (ARDI) of neoadjuvant/adjuvant FEC-100 for breast cancer. MATERIALS AND METHODS: This retrospective, single-centre cohort study including 296 patients who received FEC-100 compared PEG and non-PEG groups. The PEG group received PEG 3.6 mg as a single subcutaneous injection in each study cycle. The primary endpoint was the ARDI of FEC-100. The secondary endpoints were patient percentage of ARDI≥85%, factors associated with ARDI≥85%, and reasons for reduced ARDI. RESULTS: The PEG group showed significantly higher mean ARDI (95.6% versus 90.7%, p<0.001) and patient percentage of ARDI≥85% (93.0% versus 79.9%, p=0.001). PEG was significantly associated with ARDI≥85% (p=0.009). Neutropenia and FN, the main reasons for reduced ARDI, were significantly lower in the PEG group (p<0.05). CONCLUSION: Primary PEG 3.6 mg prophylaxis increased the ARDI of FEC-100.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Filgrastim/therapeutic use , Neoadjuvant Therapy/methods , Polyethylene Glycols/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cohort Studies , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Epirubicin/pharmacology , Epirubicin/therapeutic use , Female , Filgrastim/pharmacology , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Male , Middle Aged , Polyethylene Glycols/pharmacology , Retrospective Studies
6.
Int J Nanomedicine ; 14: 1817-1833, 2019.
Article in English | MEDLINE | ID: mdl-30880982

ABSTRACT

BACKGROUND: Gold nanorods (AuNRs), due to the optical and electronic properties namely the surface plasma resonance, have been developed to achieve the light-mediated photothermal therapy (PTT) for cancer. However, PTT alone may suffer from inefficient tumor killing. Recently, the combination of PTT and chemotherapy has been utilized to achieve synergistic anticancer effects. METHODS: In this study, AuNRs capped with hexadecyltrimethylammonium bromide (CTAB), poly(acrylic acid) (PAA), and PEGylated anisamide (a ligand known to target the sigma receptor) have been developed to produce a range of negatively charged anisamide-targeted PEGylated AuNRs (namely Au-CTAB-PAA-PEG-AA) for the combination of PTT and chemotherapy (termed as chemo-photothermal therapy [CPTT]). Epirubicin (EPI, an anthracycline drug) was efficiently loaded onto the surface of Au800-CTAB-PAA-PEG-AA via the electrostatic interaction forming Au800-CTAB-PAA-PEG-AA.EPI complex. RESULTS: The resultant complex demonstrated pH-dependent drug release, facilitated nucleus trafficking of EPI, and induced antiproliferative effects in human prostate cancer PC-3 cells. When Au800-CTAB-PAA-PEG-AA.EPI complex was further stimulated with desired laser irradiation, the synergistic outcome was evident in PC-3 xenograft mice. CONCLUSION: These results demonstrate a promising strategy for clinical application of CPTT in cancer.


Subject(s)
Drug Delivery Systems , Epirubicin/administration & dosage , Gold/chemistry , Hyperthermia, Induced , Nanotubes/chemistry , Neoplasms/therapy , Phototherapy , Polyethylene Glycols/chemistry , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzamides/chemistry , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Liberation , Endocytosis/drug effects , Epirubicin/pharmacology , Epirubicin/therapeutic use , Humans , Intracellular Space/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Nanotubes/ultrastructure , Neoplasms/drug therapy , Treatment Outcome , Xenograft Model Antitumor Assays
7.
Eur J Cancer ; 103: 184-194, 2018 11.
Article in English | MEDLINE | ID: mdl-30267987

ABSTRACT

PURPOSE: UNICANCER-PACS08 compared adjuvant FEC (5-FU; epirubicin; cyclophosphamide) then docetaxel to FEC then ixabepilone in poor prognosis early breast cancer (BC). We evaluated whether replacing docetaxel with ixabepilone would increase 5-year disease-free survival (DFS). PATIENTS AND METHODS: Triple-negative breast cancer (TNBC) or oestrogen receptor (ER)+/progesterone receptor (PR)-/HER2- BC patients were randomised to receive standard FEC (3 cycles) followed by 3 cycles of either docetaxel (100 mg/m2) or ixabepilone (40 mg/m2). Radiotherapy was mandatory after conservative surgery; ER+ patients received endocrine therapy. RESULTS: Seven hundred sixty-two patients were enrolled between October 2007 and September 2010. Baseline characteristics were balanced between arms. Median follow-up was 66.7 months. Median DFS was not reached; 5-year DFS rate was 76% with docetaxel and 79% with ixabepilone (hazard ratio [HR] = 0.80; 95% confidence interval [CI] = 0.58-1.10; p = 0.175). Median overall survival (OS) was not reached; 5-year OS rate was 86% versus 84% (HR = 0.97; 95% CI = 0.66-1.42; p = 0.897). TNBC patients treated with ixabepilone had a 23% lower risk of relapse compared to docetaxel (HR for DFS = 0.77; 95% CI = 0.53-1.11; p = 0.168). DFS was longer with ixabepilone than docetaxel in patients with grade II-III lymphocytic infiltration (HR = 0.55; 95% CI = 0.29-1.05; p = 0.063). All patients experienced ≥1 adverse events (AEs): 75% reported grade III-IV AEs and two (<1%) had grade V AEs (both with neutropenia and infection receiving ixabepilone). CONCLUSION: After adjuvant FEC, ixabepilone was comparable to docetaxel for treating poor prognosis early BC patients. The benefit of ixabepilone in subgroups (patients with TNBC and grade II-III lymphocytic infiltration) requires further evaluation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Chemotherapy, Adjuvant/methods , Cyclophosphamide/therapeutic use , Docetaxel/therapeutic use , Epirubicin/therapeutic use , Epothilones/therapeutic use , Fluorouracil/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/pathology , Cyclophosphamide/pharmacology , Docetaxel/pharmacology , Epirubicin/pharmacology , Epothilones/pharmacology , Female , Fluorouracil/pharmacology , Humans , Middle Aged , Neoplasm Staging , Survival Analysis , Young Adult
8.
Cancer Med ; 7(6): 2567-2580, 2018 06.
Article in English | MEDLINE | ID: mdl-29683262

ABSTRACT

Epirubicin is a chemotherapy agent for hepatocellular carcinoma (HCC). However, the outcome of HCC patients receiving epirubicin remains unsatisfactory. Moreover, our previous study indicated that celecoxib suppresses HCC progression and liver cancer stemness. This study evaluated the potential of celecoxib to serve as a complementary therapy during epirubicin treatment. Cell proliferation, apoptosis, invasiveness, and anchorage-independent growth were analyzed in hepatoma cells. Therapeutic efficacy was validated in rat orthotopic Novikoff hepatoma. After animal sacrifice, the antitumor mechanism of celecoxib and epirubicin combined therapy was investigated by histological analysis. Celecoxib enhanced the cytotoxic activity of epirubicin in HCC cells by promoting apoptosis. Besides, celecoxib potentiated the antineoplastic function of epirubicin in inhibiting the invasiveness and anchorage-independent growth of HCC cells. Ultrasound monitoring showed that combined therapy was more potent than either therapy alone in perturbing HCC progression. Consistently, the size and weight of dissected HCC tissues from rats receiving combined therapy were smallest among all groups. HCC treated with combined therapy exhibited the highest prevalence of apoptotic cells, which was accompanied by reduced proliferating and angiogenic activities in tumor tissues. Moreover, the expression levels of cancer stemness markers (CD44 and CD133) and drug transporter MDR-1 were significantly diminished in rats receiving combined therapy. Besides, celecoxib treatment increased the infiltration of cytotoxic T lymphocytes (CTLs) and reduced the number of regulatory T cells (Tregs), tumor-associated macrophages (TAMs), and the expression of immune checkpoint PD-L1 in HCC tissues during epirubicin therapy. Celecoxib augmented the therapeutic efficacy while modulated cancer stemness and antitumor immunity. Thus, celecoxib may serve as complementary therapy to improve the outcome of patients with advanced HCC during epirubicin treatment.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Celecoxib/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Epirubicin/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Biomarkers , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm , Drug Synergism , Humans , Immunomodulation/drug effects , Liver Neoplasms, Experimental , Rats , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(6): 772-777, 2017 Jun.
Article in Chinese | MEDLINE | ID: mdl-28615100

ABSTRACT

Objective To investigate the mechanism by which epirubicin (EPI) induces autophagy and the mechanism by which tea polyphenol (TP) regulates EPI-induced autophagy and apoptosis in T24 bladder cancer cells. Methods T24 cells weredivided into control group, EPI group, TP group and TP plus EPI group. Eight hours after corresponding treatments in different groups, transmission electron microscopy (TEM) was used to observe the image of autophagosomes. The expressions of autophagy-related protein LC3II and p62 in the cells were detected by Western blotting. Apoptotic cells were evaluated after EPI-treatment for 24 hours by flow cytometry combined with annexin V-FITC/PI staining. Western blotting was performed to determine the levels of cleaved-caspase-3 (c-caspase-3) and cleaved-PARP (c-PARP). LC3II was again tested by Western blotting 8 hours after T24 cells were treated with EPI added with autophagy pathway inhibitor chloroquine and 3-methyladenine, and moreover, the levels of LC3II and p-JNK were detected by Western blotting after T24 cells were treated with EPI combined with TP or the JNK inhibitor SP for 8 hours. Results The amount of autophagosomes and the level of LC3IIin TP plus EPI group were much lower than those in EPI group. SP reduced the level of LC3II induced by EPI. EPI increased p-JNK in a time-dependent manner. TP combined with EPI reduced the activity of JNK pathway. The apoptosis rate and the levels of c-caspase-3 and c-PARP in TP plus EPI group were much higher than those in EPI group. Conclusion TP inhibits autophagy through JNK pathway to enhance EPI-induced apoptosis in T24 bladder cancer cell line.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Epirubicin/pharmacology , Polyphenols/pharmacology , Tea , Urinary Bladder Neoplasms/pathology , Cell Line, Tumor , Humans , JNK Mitogen-Activated Protein Kinases/physiology , MAP Kinase Signaling System , Urinary Bladder Neoplasms/drug therapy
10.
Neoplasma ; 64(5): 674-680, 2017.
Article in English | MEDLINE | ID: mdl-28592119

ABSTRACT

Resistance to anticancer agents such as Epirubicin (EPI) becomes a great challenge for treating bladder cancer. However, the mechanism by which chemoresistance arised is still elusive. In the present study, we showed evidence that EPI induced cytoprotective autophagy in bladder cancer cell lines T24 and BIU87. In addition, EPI robustly activated JNK-mediated phosphorylation of Bcl-2 and disruption of Bcl-2/Beclin-1 complex. Furthermore, the green tea derivative tea polyphenol (TP) inhibited EPI-induced autophagy and promoted apoptosis induced by EPI in bladder cancer cells. These results revealed a pathway for EPI-induced autophagy that involved in JNK/Bcl-2/Beclin-1 in bladder cancer cells, and that TP synergistically promoted EPI-induced apoptosis at least partly through autophagy inhibition. Thus, TP could be utilized in combination with EPI to improve EPI-based bladder cancer therapy.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Epirubicin/pharmacology , Polyphenols/pharmacology , Tea/chemistry , Urinary Bladder Neoplasms/pathology , Beclin-1/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Urinary Bladder Neoplasms/drug therapy
11.
Asian Pac J Cancer Prev ; 17(1): 131-3, 2016.
Article in English | MEDLINE | ID: mdl-26838197

ABSTRACT

BACKGROUND: Combination chemotherapy regimes are common treatments for cancer. The aim of this study was to evaluation the effect of individual chemotherapeutic agents in comparison with a first line chemotherapy regime treatment in the AGS gastric cancer cell line by MTT assay. MATERIALS AND METHODS: In this experimental study, AGS cells were grown in RPMI-1640 supplemented with 10% fetal calf serum and 100 IU/ml penicillin, and 10 µg/ml streptomycinin, under a humidified condition at 37° with 5% CO2. All cells were washed with PBS and detached with trypsin, centrifuged and 8000 cells re-plated on to 96- well plates. LD50 doses of Epirubicin, Cisplatin and 5-fluorouracil were added to each well in mono or triple therapy. Anti-proliferative activities were determined by MTT assay after 24, 48 or 72 h. RESULTS: Results of MTT assays showed that there were no significant differences among 3 drugs in monotherapy (p=0.088), but there was significant difference between combination therapy with epirubicin (P=0.031) and 5FU (p=0.013) on cell survival at 24 h. After 48 and 72 hours, cell viability showed significant differences between the 3 drugs (p=0.048 and P=0.000 for 48 and 72 h, respectively) and there was significant difference between combination therapy with epirubicin (P=0.035 and P=0.002 for 48 and 72 h, respectively). CONCLUSIONS: The results showed no significant differences between these chemotherapy drugs each given alone, but combination therapy with 3 drugs had significant effects on cell viability in comparison with epirubicin alone.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Stomach Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Epirubicin/pharmacology , Fluorouracil/pharmacology , Humans
12.
J Bone Miner Metab ; 34(3): 277-90, 2016 May.
Article in English | MEDLINE | ID: mdl-26056019

ABSTRACT

The introduction of anthracyclines to adjuvant chemotherapy has increased survival rates among breast cancer patients. Cyclophosphamide, epirubicin and 5-fluorouracil (CEF) combination therapy is now one of the preferred regimens for treating node-positive breast cancer due to better survival with less toxicity involved. Despite the increasing use of CEF, its potential in causing adverse skeletal effects remains unclear. Using a mature female rat model mimicking the clinical setting, this study examined the effects of CEF treatment on bone and bone marrow in long bones. Following six cycles of CEF treatment (weekly intravenous injections of cyclophosphamide at 10 mg/kg, epirubicin at 2.5 mg/kg and 5-flurouracil at 10 mg/kg), a significant reduction in trabecular bone volume was observed at the metaphysis, which was associated with a reduced serum level of bone formation marker alkaline phosphatase (ALP), increased trends of osteoclast density and osteoclast area at the metaphysis, as well as an increased size of osteoclasts being formed from the bone marrow cells ex vivo. Moreover, a severe reduction of bone marrow cellularity was observed following CEF treatment, which was accompanied by an increase in marrow adipose tissue volume. This increase in marrow adiposity was associated with an expansion in adipocyte size but not in marrow adipocyte density. Overall, this study indicates that six cycles of CEF chemotherapy may induce some bone loss and severe bone marrow damage. Mechanisms for CEF-induced bone/bone marrow pathologies and potential preventive strategies warrant further investigation.


Subject(s)
Adiposity/drug effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bone Marrow Cells/metabolism , Bone Marrow/metabolism , Osteoclasts/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Marrow/pathology , Bone Marrow Cells/pathology , Cyclophosphamide/adverse effects , Cyclophosphamide/pharmacology , Epirubicin/adverse effects , Epirubicin/pharmacology , Female , Fluorouracil/adverse effects , Fluorouracil/pharmacology , Osteoclasts/pathology , Rats , Rats, Sprague-Dawley
13.
PLoS One ; 9(3): e91487, 2014.
Article in English | MEDLINE | ID: mdl-24621591

ABSTRACT

Liposomes act as efficient drug carriers. Recently, epirubicin (EPI) formulation was developed using a novel EDTA ion gradient method for drug encapsulation. This formulation displayed very good stability and drug retention in vitro in a two-year long-term stability experiment. The cryo-TEM images show drug precipitate structures different than ones formed with ammonium sulfate method, which is usually used to encapsulate anthracyclines. Its pharmacokinetic properties and its efficacy in the human breast MDA-MB-231 cancer xenograft model were also determined. The liposomal EPI formulation is eliminated slowly with an AUC of 7.6487, while the free drug has an AUC of only 0.0097. The formulation also had a much higher overall antitumor efficacy than the free drug.


Subject(s)
Breast Neoplasms/pathology , Chemistry, Pharmaceutical/methods , Edetic Acid/chemistry , Epirubicin/chemistry , Epirubicin/pharmacology , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cholesterol/chemistry , Epirubicin/administration & dosage , Epirubicin/blood , Humans , Kinetics , Liposomes , Male , Mice , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry
14.
Chem Biol Interact ; 205(3): 188-97, 2013 Oct 05.
Article in English | MEDLINE | ID: mdl-23867903

ABSTRACT

The frequent development of multidrug resistance (MDR) hampers the efficacy of available anticancer drugs in treating cervical cancer. In this study, we aimed to use formononetin (7-hydroxy-4'-methoxyisoflavone), a potential herbal isoflavone, to intensify the chemosensitivity of human cervical cancer HeLa cells to epirubicin, an anticancer drug. The reactive oxygen species (ROS) levels were correlated with MDR modulation mechanisms, including the transporter inhibition and apoptosis induction. Our results revealed that formononetin significantly enhanced the cytotoxicity of epirubicin. Co-incubation of epirubicin with formononetin increased the ROS levels, including hydrogen peroxide and superoxide free radicals. Epirubicin alone markedly increased the mRNA expression of MDR1, MDR-associated protein (MRP) 1, and MRP2. In contrast, formononetin alone or combined treatment decreased the mRNA expression of MRP1 and MRP2. This result indicates that efflux transporter-mediated epirubicin resistance is inhibited at different degrees by the addition of formononetin. This isoflavone significantly intensified epirubicin uptake into HeLa cells. Apoptosis was induced by formononetin and/or epirubicin, as signified by nuclear DNA fragmentation, chromatin condensation, increased sub-G1 and G2/M phases. The cotreatment triggered the mitochondrial apoptotic pathway indicated by increased Bax-to-Bcl-2 expression ratio, loss of mitochondrial membrane potential, and significant activation of caspase-9 and -3. In addition, extrinsic/caspases-8 apoptotic pathway was also induced by the cotreatment. N-acetyl cysteine abrogated these events induced by formononetin, supporting the involvement of ROS in the MDR reversal mechanism. This study pioneered in demonstrating that formononetin may potentiate the cytotoxicity of epirubicin in HeLa cells through the ROS-mediated MRP inhibition and concurrent activation of the mitochondrial and death receptor pathways of apoptosis. Hence, the circumvention of pump and non-pump resistance using formononetin and epirubicin may pave the way for a powerful chemotherapeutic regimen for treating human cervical cancer.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Epirubicin/pharmacology , Isoflavones/pharmacology , Phytoestrogens/pharmacology , Uterine Cervical Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antibiotics, Antineoplastic/administration & dosage , Caspases/genetics , Caspases/metabolism , Cell Survival/drug effects , Drug Synergism , Epirubicin/administration & dosage , Female , Flow Cytometry , HeLa Cells , Humans , Isoflavones/administration & dosage , Membrane Potential, Mitochondrial , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Phytoestrogens/administration & dosage , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Neoplasm/chemistry , RNA, Neoplasm/genetics , Real-Time Polymerase Chain Reaction , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
15.
Cancer Chemother Pharmacol ; 70(1): 17-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22610354

ABSTRACT

PURPOSE: Developing approaches that will increase the selectivity of anticancer drugs remains a challenge. Docosahexaenoic acid (DHA) has the potential to increase tumor sensitivity to chemotherapy with no sensitization of normal tissues. This study was aimed at exploring the mechanism involved in this differential sensitization with a focus on oxidative stress, one of the main determinants involved in DHA enhancement of anthracycline-based chemotherapy. METHODS: In a previously validated model of chemically induced mammary tumors in rats where supplemental DHA sensitized tumors to epirubicin, DHA level, lipid hydroperoxides (LPO), total antioxidant activity, glutathione peroxidase (GPx), and superoxide dismutase (SOD) activities were measured in tumors, intestine, liver, and heart in rats treated with DHA+epirubicin and in control rats (palm oil, no chemotherapy). RESULTS: At baseline, the level of LPO was similar in tumors, liver, heart, and small intestine. The proportion of DHA was higher in non-tumor tissues compared to tumors (liver or heart, p < 0.0001; intestine, p = 0.01). DHA and epirubicin induced a significant increase in LPO in tumors (p = 0.03), but no increase was detected in liver, heart, or intestine. The difference in LPO production between these tissues was associated with differences in their antioxidant defenses. In tumors, the adjustment of GPx activity was possible but limited, and no adjustment of their total antioxidant and SOD activities was observed compared to other tissues. CONCLUSION: Supplementing DHA during an anthracycline-based chemotherapy induced a selective increase of LPO in tumors. This selectivity might result from a differential handling of oxidative stress between tumor and non-tumor tissues.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Docosahexaenoic Acids/pharmacology , Epirubicin/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Oxidative Stress/drug effects , Animals , Antibiotics, Antineoplastic/administration & dosage , Antioxidants/metabolism , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/metabolism , Drug Synergism , Epirubicin/administration & dosage , Female , Glutathione Peroxidase/metabolism , Injections, Intravenous , Intestinal Mucosa/metabolism , Intestines/drug effects , Lipid Peroxides/metabolism , Liver/drug effects , Liver/metabolism , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/metabolism , Methylnitrosourea , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
16.
Med Sci Monit ; 16(10): RA218-23, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20885364

ABSTRACT

Oral ciprofloxacin might achieve higher concentration in urine than in serum; theoretically, this drug might act as an anticancer drug against bladder cancer cells. Among fluoroquinolones, ciprofloxacin is distinguished by strong inhibition of topoisomerase II. A good correlation between cytotoxic activity of ciprofloxacin toward eukaryotic cells and its ability to induce the cleavable complexes topoisomerase II-DNA has been demonstrated. These data provide a basis for supposing that ciprofloxacin may act as anticancer drug. The efforts of evaluating ciprofloxacin's influence on human bladder cell lines have been shown by many authors. The cells were exposed to ciprofloxacin at various concentrations that are attainable in the urine after oral drug administration. Antiproliferative potential of the ciprofloxacin against human bladder cells varies according to drug concentration and time of incubation. It seems that ciprofloxacin can act as an anticancer drug in eukaryotic cells. Low urine pH can enhance the antitumor effect of ciprofloxacin. Ciprofloxacin enhances the effect of action of doxorubicin and epirubicin, which are used to prevent bladder cancer recurrence after transurethral resection of superficial bladder cancer. We think that ciprofloxacin might be used for antibacterial prophylaxis and as an anticancer agent in patients with superficial bladder cancer. This idea must be checked in future placebo controlled trials.


Subject(s)
Anti-Infective Agents/therapeutic use , Carcinoma, Transitional Cell/prevention & control , Ciprofloxacin/therapeutic use , Urinary Bladder Neoplasms/prevention & control , Antineoplastic Agents/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Cell Line, Tumor , Ciprofloxacin/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Drug Screening Assays, Antitumor , Drug Therapy, Combination , Epirubicin/administration & dosage , Epirubicin/pharmacology , Epirubicin/therapeutic use , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/prevention & control , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology
17.
Breast Cancer Res Treat ; 113(2): 231-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18278552

ABSTRACT

Gene expression data has in recent years demonstrated the superior capacity to predict the prognosis of breast cancer patients unreceiving adjuvant chemotherapy comparing to the information available from traditional clinical and pathological sources. Meanwhile, adjuvant chemotherapy can significantly improve survival of breast cancer. It would be inappropriate to ignore its effect on prognosis. We hypothesized that an integrated gene expression profile can predict the prognosis of breast cancer patients receiving chemotherapy. Therefore, we screened the specific gene markers and constructed an integrated 24-gene signature by low-density microarray including the "poor signature" and genes related to resistance to chemotherapy. The gene signature stratified correctly patients into good prognosis group and poor prognosis group. In addition, the Kaplan-Meier analyses for disease-free survival as a function of the 24-gene signature showed highly significant differences between the two groups (Log Rank test P < 0.0001 = Univariate and multivariate Cox's proportional-hazards regression analyses indicated that the signature represents the strongest independent prognostic factor for breast cancer patients. When compared with single signature, such as Oncotype DX and 70 poor signature, the integrated signature showed more predominant power of predication in breast cancer patients receiving chemotherapy. Such integrated signature will critically aid clinical decision making at the level of individualization for most breast cancer patients receiving chemotherapy.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Chemotherapy, Adjuvant , Gene Expression Profiling/methods , Genes, Neoplasm , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Carcinoma/chemistry , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/mortality , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/mortality , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacology , Disease-Free Survival , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Epirubicin/administration & dosage , Epirubicin/pharmacology , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Follow-Up Studies , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kaplan-Meier Estimate , Middle Aged , Prognosis , Proportional Hazards Models , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Young Adult
18.
Nat Genet ; 40(7): 844-53, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18511948

ABSTRACT

NQO1 guards against oxidative stress and carcinogenesis and stabilizes p53. We find that a homozygous common missense variant (NQO1(*)2, rs1800566(T), NM_000903.2:c.558C>T) that disables NQO1 strongly predicts poor survival among two independent series of women with breast cancer (P = 0.002, N = 1,005; P = 0.005, N = 1,162), an effect particularly evident after anthracycline-based adjuvant chemotherapy with epirubicin (P = 7.52 x 10(-6)) and in p53-aberrant tumors (P = 6.15 x 10(-5)). Survival after metastasis was reduced among NQO1(*)2 homozygotes, further implicating NQO1 deficiency in cancer progression and treatment resistance. Consistently, response to epirubicin was impaired in NQO1(*)2-homozygous breast carcinoma cells in vitro, reflecting both p53-linked and p53-independent roles of NQO1. We propose a model of defective anthracycline response in NQO1-deficient breast tumors, along with increased genomic instability promoted by elevated reactive oxygen species (ROS), and suggest that the NQO1 genotype is a prognostic and predictive marker for breast cancer.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , Polymorphism, Single Nucleotide , Antineoplastic Agents/therapeutic use , Antioxidants/metabolism , Antioxidants/physiology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Death/genetics , Combined Modality Therapy , Drug Resistance, Neoplasm/genetics , Epirubicin/pharmacology , Female , Follow-Up Studies , Genotype , Homozygote , Humans , Models, Biological , NAD(P)H Dehydrogenase (Quinone)/physiology , Neoplasm Metastasis , Prognosis , Survival Analysis , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
19.
Cancer Lett ; 252(2): 195-207, 2007 Jul 18.
Article in English | MEDLINE | ID: mdl-17275999

ABSTRACT

One of the cornerstones of therapy for invasive breast cancer includes the use of anthracyclines. Epirubicin, a stereoisomer of doxorubicin, is one of the commonly used anthracyclines. Anthracyclines while effective therapy for breast cancer, have their own unique toxicities, such as cardiomyopathy. l-Carnitine, a quarternary ammonium compound synthesized from methionine and lysine, is required for oxidative metabolism in mitochondria. Cardiac function is closely linked with oxidative metabolism whereby l-carnitine is an essential cofactor. A hypothesis is being investigated to determine if supplementation with carnitine in breast cancer patients treated with epirubicin will reduce the development of cardiac toxicity. We determined whether addition of l-carnitine altered the tumor cytotoxic effects of epirubicin using a number of in vitro cell viability assays in different breast cancer cell lines including BT549, MDA-MB-435, NCI-ADR-RES, MCF7 and T47D. Additionally we investigated the ability of cells to respond to l-carnitine following analysis of the expression of carnitine metabolic enzymes by RT-PCR. We determined that supplementation with l-carnitine had no effect on the ability of epirubicin to kill a variety of breast cancer cell lines. Additionally, no differences in the induction of apoptosis by epirubicin were observed. Furthermore, all cell lines examined expressed proteins required for carnitine uptake and use. Our data suggest that supplementation with l-carnitine does not impair the ability of epirubicin to kill breast cancer cells. These results suggest that supplementation with l-carnitine in patients undergoing epirubicin treatment could be safely used to reduce associated cardiotoxicities without fear that the efficacy of chemotherapy is jeopardized.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/pathology , Carnitine/pharmacology , Epirubicin/pharmacology , Base Sequence , Blotting, Western , Carnitine/administration & dosage , Cell Line, Tumor , DNA Primers , Humans , Reverse Transcriptase Polymerase Chain Reaction
20.
Eur Urol ; 51(4): 962-9; discussion 969-70, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17084017

ABSTRACT

OBJECTIVES: Three-dimensional (3D) spheroids are a good model for studying in vitro chemosensitivity because they reproduce unicellular and multicellular mechanisms of drug resistance. We aimed to develop a chemosensitivity test for intravesical drugs and to also verify the effects of verapamil (VPM) and ciprofloxacin (CIPRO). METHODS: Cold cup biopsies from 40 superficial bladder tumours were taken, fragmented, and left in culture. 3D-spheroids were obtained and transferred into a 24 multiwell dish containing (1) wells 1-3: 1 mg/ml epirubicin (EPI); (2) 4-6: 1 mg/ml EPI+0.5 mg/ml VPM; (3) 7-9: 1 mg/ml adriamycin (ADR); (4) 10-12: 1 mg/ml thiotepa (THIO); (5) 13-15: 1 mg/ml mitomycin C (MMC); (6) 16-18: 1mg/ml EPI+0.2 mg/ml CIPRO; (7) 19-21: 0.2 mg/ml CIPRO; (8) 22-24: controls. Sensitivity was calculated by using the trypan blue assay. RESULTS: Evaluability of clinically relevant tests (G1-G2 lesions) was 84% (21 of 25 patients). MMC was the best agent (p<0.001) with mean sensitivity being 50%, followed by THIO (37%), EPI (7%), and ADR (3%). We found no significant difference (p=0.370) between CIPRO and the control, or between EPI+CIPRO and EPI alone (p=0.550). VPM markedly enhanced sensitivity to EPI compared with EPI alone (97% vs. 7%, p<0.001). CONCLUSIONS: Our assay allows determining sensitivity to several drugs in superficial bladder tumours. It might be used in clinical practice to select the best drug for each patient. It also has experimental utility in investigating the effect of new drugs or combinations. VPM reverted resistance to EPI. CIPRO showed no effect on bladder tumour spheroids.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Spheroids, Cellular/drug effects , Urinary Bladder Neoplasms/drug therapy , Ciprofloxacin/pharmacology , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Epirubicin/pharmacology , Epirubicin/therapeutic use , Humans , Mitomycin/pharmacology , Mitomycin/therapeutic use , Thiotepa/pharmacology , Thiotepa/therapeutic use , Tumor Cells, Cultured , Urinary Bladder Neoplasms/pathology , Verapamil/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL