Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
1.
Medicine (Baltimore) ; 100(14): e25398, 2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33832134

ABSTRACT

OBJECTIVES: To assess the efficacy and toxicity of gemcitabine-based induction chemotherapy followed by concurrent chemoradiotherapy (CCRT) in locally advanced nasopharyngeal carcinoma (LA-NPC). METHODS: Both observational studies (OBS) and randomized controlled trials (RCT) were included in the meta-analysis. Systematic online searches were conducted in Web of Sciences, PubMed, Embase, meeting proceedings and ClinicalTrials.gov from the inception to May 25, 2020. The primary endpoint of interest was overall survival. RESULTS: five OBSs and 2 RCTs including 1680 patients were incorporated in the analysis. The evidence from the RCTs showed that adding gemcitabine-based induction chemotherapy to CCRT significantly improved progression free survival (hazard ratio (HR): 0.60, 95% confidence interval (CI): 0.40-0.88; P = .010; chi square P = .25; I2 = 24%) and overall survival (HR: 0.47; 95% CI: 0.28-0.80; P = 0.005; chi square P = .49, I2 = 0%) and was related to a higher risk of hematological toxicities. Furthermore, based on the data of OBSs, overall survival (HR: 0.52; 95% CI: 0.31-0.88; P = .02; chi square P = .37, I2 = 6%) was significantly improved in patients treated with gemcitabine-based induction chemotherapy compared to those treated with taxane-based induction chemotherapy. However, the progression free survival (HR: 0.67; 95% CI: 0.45-1.01; P = .06; chi square P = .74; I2 = 0%) showed no significant difference. CONCLUSIONS: For LA-NPC patients, adding gemcitabine-based induction chemotherapy to CCRT significantly improved overall survival and progression free survival with a higher risk of hematological toxicities when compared to CCRT alone. Also, gemcitabine-based regimen could be used as an alternative induction chemotherapy regimen to taxane-based regimen in the treatment of LA-NPC.


Subject(s)
Deoxycytidine/analogs & derivatives , Induction Chemotherapy/methods , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Neoplasms/pathology , Antimetabolites, Antineoplastic/therapeutic use , Antimetabolites, Antineoplastic/toxicity , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/toxicity , Bridged-Ring Compounds/therapeutic use , Bridged-Ring Compounds/toxicity , Case-Control Studies , Chemoradiotherapy/methods , China/epidemiology , Combined Modality Therapy/methods , Deoxycytidine/therapeutic use , Deoxycytidine/toxicity , Humans , Induction Chemotherapy/trends , Middle Aged , Nasopharyngeal Carcinoma/mortality , Nasopharyngeal Carcinoma/radiotherapy , Neoplasm Staging , Observational Studies as Topic , Progression-Free Survival , Randomized Controlled Trials as Topic , Survival Analysis , Taxoids/therapeutic use , Taxoids/toxicity , Treatment Outcome , Gemcitabine
2.
Hum Exp Toxicol ; 40(7): 1122-1129, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33380212

ABSTRACT

BACKGROUND: We evaluated the effect of cabazitaxel (CAB) as a third-line taxane on Toll-like receptor 4 (TLR4)-mediated signaling pathways, especially NF-κB activity, in metastatic castration-resistant prostate cancer (mCRPC) cells. METHODS: CAB cytotoxicity was determined by WST-1 assay. To assess the relationship between CAB efficacy and TLR4 signaling pathways, RT-PCR, western blot and immunofluorescence analysis were performed. Additionally, CAB-mediated apoptotic cell death was assessed by Annexin V and RT-PCR analysis. RESULTS: Our results demonstrated that CAB exerted considerably cytotoxic and apoptotic effects on PC-3 mCRPC cells (p < 0.05). CAB treatment altered TLR4 expression level in a dose-dependent manner. Furthermore, 1 nM CAB treatment significantly induced NF-κB activity through p65 nuclear localization and increased the expression level of caspase-3, Bax and p53. Interestingly, total apoptotic cell death and IRF3 protein levels were increased at 5 nM concentration of CAB despite a decrease in the levels of both NF-κB and pro-apoptotic genes. CONCLUSIONS: Therefore, NF-κB activity may be a potential target for the efficacy of CAB in mCRPC cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Prostatic Neoplasms, Castration-Resistant/drug therapy , Signal Transduction/drug effects , Taxoids/toxicity , Taxoids/therapeutic use , Toll-Like Receptor 4/therapeutic use , Cell Survival/drug effects , Cells, Cultured/drug effects , Humans , Male , Prostatic Neoplasms, Castration-Resistant/physiopathology , Treatment Outcome
3.
Trials ; 21(1): 391, 2020 May 07.
Article in English | MEDLINE | ID: mdl-32381018

ABSTRACT

BACKGROUND: Trastuzumab (Tmab), pertuzumab (Pmab), and taxane has been a standard first-line treatment for recurrent or metastatic human epidermal growth factor (HER2)-positive breast cancer (HER2+ mBC) but has some safety issues due to taxane-induced toxicities. This has led to ongoing efforts to seek less toxic alternatives to taxanes that are equally effective when used in combination with Tmab plus Pmab. This study aims to show the non-inferiority of eribulin, a non-taxane microtubule inhibitor, against taxane, as a partner for dual HER2 blockade. METHODS/DESIGN: This multicenter, randomized, open-label, parallel-group, phase III study will involve a total of 480 Japanese women with HER2+ mBC who meet the following requirements: (1) age 20-70 years; (2) no prior cytotoxic chemotherapy (excluding trastuzumab-emtansine) for mBC; (3) ≥ 6 months after prior neoadjuvant or adjuvant cytotoxic chemotherapy; (4) presence of any radiologically evaluable lesion; (5) left ventricular ejection fraction ≥ 50%; (6) Eastern Cooperative Oncology Group performance status score of 0 or 1; (7) adequate organ function; and (8) life expectancy of at least 6 months. They will be randomized 1:1 to receive eribulin (1.4 mg/m2 on days 1 and 8) or taxane (docetaxel 75 mg/m2 on day 1 or paclitaxel 80 mg/m2 on days 1, 8, and 15) in combination with Tmab (8 mg/kg then 6 mg/kg) plus Pmab (840 mg then 420 mg) on day 1 of each 21-day cycle. The treatment will be continued until disease progression or unmanageable toxicity. The primary endpoint is progression-free survival as per investigator according to RECIST v1.1 criteria. Key secondary endpoints include objective response rate, overall survival, quality of life and safety. Non-inferiority will be tested with two margins of 1.33 and 1.25 in a stepwise manner. If non-inferiority is shown with a margin of 1.25, superiority will then be tested. DISCUSSION: If this study shows the non-inferiority, or even superiority, of Tmab, Pmab, and eribulin against the existing taxane-containing regimen, this new regimen may become a standard first- or second-line treatment option for HER2+ mBC in Japan. TRIAL REGISTRATION: ClinicalTrials.gov, ID: NCT03264547. Registered on 28 June 2017.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/secondary , Neoplasm Metastasis/drug therapy , Receptor, ErbB-2/metabolism , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/pathology , Bridged-Ring Compounds/administration & dosage , Bridged-Ring Compounds/therapeutic use , Bridged-Ring Compounds/toxicity , Female , Furans/administration & dosage , Furans/therapeutic use , Humans , Japan/epidemiology , Ketones/administration & dosage , Ketones/therapeutic use , Middle Aged , Progression-Free Survival , Quality of Life , Stroke Volume/physiology , Taxoids/administration & dosage , Taxoids/therapeutic use , Taxoids/toxicity , Trastuzumab/administration & dosage , Trastuzumab/therapeutic use , Tubulin Modulators/administration & dosage , Tubulin Modulators/therapeutic use , Ventricular Function, Left/physiology
4.
Clin Neurophysiol ; 131(8): 1979-1985, 2020 08.
Article in English | MEDLINE | ID: mdl-32291143

ABSTRACT

OBJECTIVE: To comprehensively describe patient-reported, functional and neurophysiological outcomes to elucidate the phenotypic profile of taxane-induced neuropathy. METHODS: Taxane-treated patients (n = 47) completed cross-sectional bilateral clinical and sensory assessments and nerve conduction studies. Patients reported symptom severity via Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (FACT/GOG-Ntx13) questionnaire. RESULTS: Symptoms of neuropathy were reported by 81% of patients. On clinical examination, 62% had 2 or more abnormalities, with 20% indicating significant symptomatic and objective neuropathy. Nerve conduction studies were consistent with a sensory predominant axonal neuropathy. However, features more typical of entrapment neuropathy were also present in > 50%, which were not associated with overall severity of chemotherapy-induced peripheral neuropathy (CIPN) or clinical risk factors. CONCLUSIONS: There is considerable variation in CIPN phenotypes associated with taxane-treatment. Understanding their clinical associations may assist in identification of patients at risk of severe neurotoxicity. This would enable treatment modification decisions but also limit early cessation of effective anti-cancer treatment in patients with less severe neurological sequelae. SIGNIFICANCE: Understanding the CIPN phenotype may inform treatment decisions which could impact clinical and survival outcomes.


Subject(s)
Antineoplastic Agents/toxicity , Bridged-Ring Compounds/toxicity , Electrodiagnosis/methods , Neurotoxicity Syndromes/physiopathology , Peripheral Nervous System Diseases/physiopathology , Phenotype , Taxoids/toxicity , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Bridged-Ring Compounds/administration & dosage , Bridged-Ring Compounds/therapeutic use , Electrodiagnosis/standards , Female , Humans , Middle Aged , Neurotoxicity Syndromes/diagnosis , Neurotoxicity Syndromes/etiology , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/etiology , Taxoids/administration & dosage , Taxoids/therapeutic use , Urogenital Neoplasms/drug therapy
5.
Cancer Treat Res Commun ; 15: 1-6, 2018.
Article in English | MEDLINE | ID: mdl-30207281

ABSTRACT

MICROABSTRACT: Women treated with chest radiation for Hodgkin lymphoma (HL) have significantly higher risk of developing breast cancer, and little is known about how these patients tolerate chemotherapy for breast cancer. This small retrospective study identified 15 patients, noting that these patients tolerate proposed chemotherapy regimens for breast cancer in rates similar to those without prior HL and therapeutic radiation. PURPOSE: Women treated for Hodgkin lymphoma (HL) with chest radiation have significantly higher risk of developing breast cancer, and little is known about how these patients tolerate chemotherapy for breast cancer. METHODS: Women with breast cancer diagnosed from 1986-2015 after radiation for HL were identified from hospitals and clinics in St. Paul and Minneapolis, Minnesota. Patient, tumor and treatment characteristics, and clinical outcomes were abstracted from medical records and summarized using descriptive statistics. Chemotherapy was defined as tolerated if all scheduled doses and cycles were completed without deviation from the initial plan, with lack of grade 3 or higher toxicity attributable to chemotherapy in categories including blood, cardiac, gastrointestinal, fatigue and pain. RESULTS: Forty-two patients with breast cancer and prior radiation for HL were identified, 15 of which received chemotherapy for breast cancer. We noted 75% tolerability of taxane-based and 100% tolerability of anthracycline-based chemotherapy, suggesting that most patients with prior radiation for HL tolerate chemotherapy for breast cancer. A subset of patients (N = 7) in this study were also treated with chemotherapy for HL prior to breast cancer diagnosis, and 86% (6 of 7) also tolerated chemotherapy for breast cancer. CONCLUSIONS: Treatment of breast cancer is strongly influenced by prior treatment of HL. Although this study was small and did not meet statistical significance, the data suggest that these patients tolerate proposed chemotherapy regimens for breast cancer in rates similar to those without prior HL and therapeutic radiation. Larger studies comparing specific chemotherapy dosing schedules are needed to address this complicated population.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/toxicity , Breast Neoplasms/drug therapy , Hodgkin Disease/radiotherapy , Neoplasms, Radiation-Induced/drug therapy , Neoplasms, Second Primary/drug therapy , Adult , Anthracyclines/administration & dosage , Anthracyclines/therapeutic use , Anthracyclines/toxicity , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Breast Neoplasms/etiology , Bridged-Ring Compounds/administration & dosage , Bridged-Ring Compounds/therapeutic use , Bridged-Ring Compounds/toxicity , Cancer Survivors/statistics & numerical data , Cohort Studies , Female , Humans , Minnesota , Neoplasms, Radiation-Induced/etiology , Neoplasms, Second Primary/etiology , Pilot Projects , Radiotherapy/adverse effects , Retrospective Studies , Taxoids/administration & dosage , Taxoids/therapeutic use , Taxoids/toxicity
6.
Molecules ; 23(8)2018 Jul 24.
Article in English | MEDLINE | ID: mdl-30042342

ABSTRACT

Previous studies of the neuroprotective activity of polyphenols have used ununiform culture systems, making it difficult to compare their neuroprotective potency. We have established a new and simple method for preparing differentiated PC12 cells by removing the toxic coating step. Cells were induced to differentiate with the nerve growth factor (NGF) in a serum-free medium, without a medium change, but with a one-time overlay supplementation of NGF. The optimal inoculation density of the cells was 6⁻12 × 10³ cells/cm², and the presence of serum inhibited the differentiation. Neuroprotective activity could be quantified by the specific index (SI) value, that is, the ratio of the 50% cytotoxic concentration to the 50% effective concentration. Alkaline extract from the leaves of Sasa senanensis Rehder (SE), having had hormetic growth stimulation, showed the highest SI value, followed by epigallocatechin gallate. The SI value of curcumin and resveratrol was much lower. This simple overly method, that can prepare massive differentiated neuronal cells, may be applicable for the study of the differentiation-associated changes in intracellular metabolites, and the interaction between neuronal cells and physiological factors.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Cell Culture Techniques , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Sasa/chemistry , Taxoids/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Animals , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Differentiation/drug effects , Culture Media, Serum-Free/pharmacology , Curcumin/pharmacology , Hormesis , Nerve Growth Factor/pharmacology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/isolation & purification , PC12 Cells , Plant Extracts/isolation & purification , Plant Leaves/chemistry , Polyphenols/isolation & purification , Polyphenols/pharmacology , Rats , Resveratrol , Stilbenes/pharmacology , Taxoids/toxicity
7.
Prostate ; 78(12): 905-914, 2018 09.
Article in English | MEDLINE | ID: mdl-29749077

ABSTRACT

BACKGROUND: Taxanes chemotherapies represent the major therapeutic alternative for symptomatic mCRPC. While docetaxel is the most commonly prescribed Taxane for mCRPC; cabazitaxel has been approved for patients unresponsive to docetaxel. Still mCRPC remains incurable and patients often experience severe side effects. Recently, the FIRSTANA trial first demonstrated the absence of superiority in overall survival between cabazitaxel and docetaxel in mCRPC patients. Inversely, different toxicity were reported suggesting that cabazitaxel may provide a first line treatment option for some patients urging for a deeper characterization of cabazitaxel mechanisms of action as well as a re-evaluation of cabazitaxel conventional dose and schedule. In this study, our goal was therefore to evaluate the anti-tumor efficacy of various cabazitaxel regimens delivered as monotherapy or in combination with PEDF, a known anti-angiogenic and anti-neoplastic agent. METHODS: CRPC cells undergoing Taxane treatment were evaluated for cell proliferation, migration and death, and apoptosis using crystal violet staining, chemotaxis, cell cycle, and TUNEL assays. In vitro data were corroborated in CL1 CRPC xenografts where mice received intermittent or metronomic low-doses cabazitaxel ± PEDF. RESULTS: We found that cabazitaxel inhibits the proliferation of CRPC cells with a higher efficacy than docetaxel in vitro. As expected, high-doses of Taxanes blocked the cells in mitosis. Surprisingly, low-doses of cabazitaxel induced more cell death than docetaxel mainly through apoptosis. In vivo, intermittent cabazitaxel lead to disease stabilization when combined with PEDF. Unexpectedly, low-doses of cabazitaxel delayed tumor growth with severe toxicity for some of the doses tested. Other results showed that PEDF and low-doses of cabazitaxel combination inhibited the migration of tumor cell and increased the tumoricidal activity of macrophages toward prostate tumor cells. CONCLUSIONS: Our findings highlight the great promise of cabazitaxel drug and predict a possible move of cabazitaxel forward within the therapeutic sequence of prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Eye Proteins/pharmacology , Neoplasm Metastasis/drug therapy , Nerve Growth Factors/pharmacology , Prostatic Neoplasms, Castration-Resistant/pathology , Serpins/pharmacology , Taxoids/pharmacology , Animals , Apoptosis/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Docetaxel/therapeutic use , Docetaxel/toxicity , Dose-Response Relationship, Drug , Humans , Macrophages/drug effects , Macrophages/physiology , Male , Mice , Mice, SCID , PC-3 Cells , Recombinant Proteins/pharmacology , Taxoids/therapeutic use , Taxoids/toxicity , Xenograft Model Antitumor Assays
8.
Metas enferm ; 21(3): 67-73, abr. 2018. tab, graf
Article in Spanish | IBECS | ID: ibc-172676

ABSTRACT

OBJETIVO: determinar la incidencia y evolución de la neuropatía periférica inducida por taxanos (NPIT) en pacientes con cáncer de mama y valorar la influencia de la NPIT en la calidad de vida global (CV). MÉTODO: estudio descriptivo longitudinal prospectivo realizado en el Servicio de Oncología del Hospital Clínic de Barcelona (julio 2015-abril 2016). Se incluyeron mujeres diagnosticadas de cáncer de mama en su primera línea de tratamiento con quimioterapia (paclitaxel o docetaxel) en quienes se evaluó la neurotoxicidad y la calidad de vida mediante tres cuestionarios autoinformados validados al inicio, a las 6 y 12 semanas y un mes tras finalizar el tratamiento. RESULTADOS: participaron 33 pacientes de las cuales el 84,4% había desarrollado algún grado de NP al final del seguimiento. La neurotoxicidad empeoró de manera estadísticamente significativa con la acumulación de dosis hasta el final del tratamiento y se mantuvo estable un mes tras la última administración (p< 0,001). La neuropatía sensitiva aumentó de manera estadísticamente significativa a lo largo del seguimiento (p< 0,001). La afectación de la sensibilidad motora también, salvo en la última medición (p< 0,005). Se observó una correlación positiva entre la neurotoxicidad y deterioro de la CV (r= 0,609 (p< 0,0001)) CONCLUSIONES: la NPIT es un efecto secundario con una alta incidencia en la población de mujeres con cáncer de mama estudiada y provoca un efecto negativo en la CV percibida de las pacientes. Las enfermeras oncológicas son profesionales clave en la prevención y el manejo de este efecto secundario


OBJECTIVE: to determine the incidence and evolution of taxane-induced peripheral neuropathy (TIPN) in breast cancer patients, and to assess the influence of TIPN on overall quality of life (QoL). METHOD: a prospective longitudinal descriptive study conducted at the Oncology Unit of the Hospital Clínic de Barcelona (July, 2015- April, 2016). The study included women with diagnosis of breast cancer, on their first line of treatment with chemotherapy (paclitaxel or docetaxel); neurotoxicity and quality of life were evaluated through three self-reported questionnaires validated at baseline, at 6 and 12 weeks, and one month after completing treatment. RESULTS: the study included 33 patients; 84.4% of them had developed some degree of PN at the end of follow-up. There was a statistically significant worsening in neurotoxicity with dose accumulation until the end of the treatment, and it remained stable one month after the last administration (p< 0.001). There was a statistically significant increase in sensitive neuropathy throughout follow-up (p< 0.001); also in terms of involvement in motor sensitivity, except in the final measurement (p< 0-005). A positive correlation was observed between neurotoxicity and QoL deterioration (r= 0.609 (p < 0.0001)). CONCLUSIONS: TIPN is a side effect with high incidence among the population studied of women with breast cancer, and it causes a negative impact on patient-perceived QoL. Oncology nurses are the key professionals for the prevention and management of this side effect


Subject(s)
Humans , Female , Adult , Middle Aged , Aged , Breast Neoplasms/drug therapy , Antineoplastic Agents/adverse effects , Taxoids/toxicity , Prospective Studies , Breast Neoplasms/complications , Neurotoxins/adverse effects , Quality of Life , Paclitaxel/toxicity , Treatment Outcome , Self Report
9.
Biomed Pharmacother ; 97: 1000-1010, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29136767

ABSTRACT

To take full advantage of combination therapy of Docetaxel (DTX) and Magnesium isoglycyrrhizinate (MGIG), the pharmacokinetic- pharmacodynamic- toxicodynamics (PK-PD-TD) interaction of DTX and MGIG in non-small cell lung tumor-bearing mice was investigated in the present study. A model, an integrated semi-mechanistic PK-PD-TD, was established for elucidating the exposure-effect-toxicity relationship between DTX and MGIG. A tumor growth and a transit compartmental system were applied to imitate the growth and death of tumor cell. An indirect model with precursor-dependence was induced to clarify the temporal relationship between liver injury and drug exposure. No PK interaction between DTX and MGIG in plasma, liver and tumor was observed. In PD and TD results, MGIG had no antitumoral activity on non-small cell lung carcinoma, while it showed a strong hepatoprotection on DTX-induced liver injury. The PK-PD parameters of anti-tumor effect were related with the tumor growth characteristics, the kinetics of the tumor death and drug potency. In the PK-TD model, it was estimated about the elevation rate of ALT after DTX challenge in hepatocytes as well as plasma. MGIG reduced the DTX-induced ALT release rate from hepatocyte efficiently. Based on parameters estimated via PK-PD-TD correlation, the model successfully predicted the tumor growth kinetics and hepatoprotection at different dose regimes. Therefore, this prospective model might provide an alternative approache to the optimization of new experiment design.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Saponins/pharmacology , Taxoids/pharmacology , Triterpenes/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Carcinoma, Non-Small-Cell Lung/pathology , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/prevention & control , Docetaxel , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Models, Biological , Saponins/administration & dosage , Saponins/toxicity , Taxoids/administration & dosage , Taxoids/toxicity , Triterpenes/administration & dosage , Triterpenes/toxicity , Xenograft Model Antitumor Assays
10.
Curr Drug Deliv ; 15(4): 564-575, 2018.
Article in English | MEDLINE | ID: mdl-28637416

ABSTRACT

BACKGROUND: Docetaxel (DTX) has been used to treat several types of cancers, but it has provided pharmaceutical challenges due to its poor water solubility and toxicities associated with the co-solvents (tween-80 and ethanol). Nanopolymer therapeutics can be engineered to deliver anticancer agent specifically to cancer cells, thereby leaving normal healthy cells unaffected by toxic drugs such as DTX. The objective of the present study was to synthesize the polyacrylic acid (PAA)-DTX conjugate (PAADC) and preparation of nanopolymer therapeutics such as PAADC/DSPE-mPEG2000 mixed micelles (PAADC-DP MMs). METHODS: The prepared PAADC-DP MMs were characterized for mean particle size and zeta potential, in vitro release profile using dialysis technique, hemolytic behavior against human blood, and cytotoxicity against human cancer cell line (A549) using MTT assay. In vivo acute toxicity of PAADC-DP MMs was determined in albino mice at intravenous single dose of 40 mg/kg. RESULTS: PAADC-DP MMs showed mean particle size of 443±9nm. PAADC-DP MMs showed maximum DTX loading (DTX equivalent; 90.5±2.7%) with minimum DSPE-mPEG2000 molecules (1:1 ratio), while to load 77.9±2.2% of plain DTX, more DSPE-mPEG2000 is required(1:10 ratio). The developed PAADC-DP MMs system showed significantly lower CMC (5 ng/mL), sustained release profile (28.6±1.9% after 48 h of study), lower hemolytic behavior (13.7±1.3% of hemolysis ratio at 40 µg/mL concentration and after 1 h incubation), higher in vitro cytotoxicity (IC50 of 0.0064±0.001 nM after 48 h study) and remarkably reduced in vivo toxicity (9.9±2.1% body weight loss) in mice when compared to marketed Taxotere®. CONCLUSION: The obtained results clearly demonstrated that the developed PAADC-DP MMs system is a promising approach for cancer chemotherapy with reduced toxicity.


Subject(s)
Cell Survival/drug effects , Drug Delivery Systems/methods , Micelles , Polymers/chemistry , Taxoids/administration & dosage , Taxoids/chemistry , Acrylic Resins/chemistry , Animals , Cell Line, Tumor , Docetaxel , Drug Liberation , Female , Hemolysis/drug effects , Humans , Mice , Nanotechnology/methods , Particle Size , Solubility , Surface Properties , Taxoids/pharmacology , Taxoids/toxicity
11.
Cancer Lett ; 410: 169-179, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28965854

ABSTRACT

Effective treatment of metastatic castration resistant prostate cancer (mCRPC) remains an unmet challenge. Cabazitaxel (CBZ) is approved for mCRPC after docetaxel (DTX) failure, but the improvement in survival is only moderate (∼2 months) and patients suffer from significant side effects. Here, we report the development of a polymer based delivery system for CBZ to improve its safety and efficacy against DTX-resistant mCRPC. CBZ was conjugated to a carboxymethylcellulose-based polymer (Cellax-CBZ), which self-assembled into ∼100 nm particles in saline and exhibited sustained drug release in serum at 10%/day. Cellax-CBZ delivered 157-fold higher CBZ to PC3-RES prostate tumor in mice and could be safely administered at a 25-fold higher dose compared to free CBZ, resulting in superior tumor inhibition in multiple mice models of DTX-resistant CRPC. In a metastatic bone model of CRPC, Cellax-CBZ significantly improves overall survival with a 70% long-term survival rate to day 120, while mice treated with free CBZ had a median survival of 40 days. Cellax-CBZ induced mild and reversible neutropenia in mice but no other tissue damage. Cellax-CBZ showed significant potential for improving therapy of mCRPC over clinically approved CBZ.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Drug Resistance, Neoplasm , Nanoparticles , Prostatic Neoplasms, Castration-Resistant/drug therapy , Taxoids/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Bone Neoplasms/secondary , Carboxymethylcellulose Sodium/chemistry , Cell Line, Tumor , Delayed-Action Preparations , Docetaxel , Drug Carriers , Drug Compounding , Drug Liberation , Humans , Male , Maximum Tolerated Dose , Mice, Inbred NOD , Mice, SCID , Neutropenia/chemically induced , Particle Size , Prostatic Neoplasms, Castration-Resistant/pathology , Solubility , Taxoids/chemistry , Taxoids/pharmacokinetics , Taxoids/toxicity , Tissue Distribution , Xenograft Model Antitumor Assays
12.
Chem Pharm Bull (Tokyo) ; 65(10): 959-966, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28781335

ABSTRACT

The application of chemotherapeutics with chemical drugs is always challenged by their high toxicities throughout the body in clinical trials. Here, we reported a smart formulation of docetaxel developed by solid dispersion and effervescent techniques for efficient and safe delivery of chemical drug to lung tissue. To achieve a high delivery to lung with reduced systemic toxicity, docetaxel was loaded into a kind of lecithoid nanoparticles (DTX-LN) which were prepared by a solid dispersion and effervescent method. After intravenous administration of DTX-LN to rabbit, the docetaxel level in lung was approximately 37-fold higher than that of docetaxel injection (DTX-INJ, a commercial injection preparation of DTX/polysorbate 80 micelles) group at 0.5 h and showed the highest tissue distribution among all the organs. Besides, the targeting parameter Re value of total increased amount of DTX in lung (AUC0-t) ratio (DTX-LN to DTX-INJ) is about 16.69, indicating a significantly enhanced lung targeting ability of DTX-LN. In subacute toxicity study, DTX-LN displayed a reduced hematotoxicity, especially for the negative impacts on white blood cells, lymphocyte and granulocyte when compared with DTX-INJ during both weekly and 3-weekly schedules administration. In addition, histopathological analysis demonstrated that DTX-LN showed less tissue damages on rabbit heart and kidney compared to DTX-INJ. Hence, this work would provide an insight for improving lung delivery efficacy of drugs with reduced systemic toxicity.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Taxoids/chemistry , A549 Cells , Animals , Cell Survival/drug effects , Docetaxel , Hemolysis/drug effects , Humans , Lung/drug effects , Lung/metabolism , Micelles , Models, Animal , Particle Size , Polysorbates/chemistry , Rabbits , Taxoids/pharmacokinetics , Taxoids/toxicity , Tissue Distribution
13.
J Physiol Pharmacol ; 68(2): 295-308, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28614779

ABSTRACT

The anticancer drugs doxorubicin (DOX), paclitaxel (PTX) and docetaxel (DTX) have been proven to induce oxidative stress (OS)-dependent side-effects in non-targeted tissues. In normal conditions, the blood-brain barrier (BBB) prevents these drugs from penetrating into the brain. However, some studies have demonstrated that small amounts of DOX can penetrate the brain via an oxidatively impaired BBB and cause damage, which suggests that including antioxidants in chemotherapy could possibly protect the brain against the toxicity of anticancer drugs. We investigated whether DOX, DTX and PTX can induce oxidative damage in rat brains in vivo and whether inclusion of the nitroxyl antioxidant Pirolin (PL) to DOX/taxane chemotherapy can protect the brain from the OS toxicity of these drugs. Wistar rats received i.p. a single dose (10 mg/kg b.w.) of DOX, DTX, PTX or PL alone or a combination of a drug + PL. After four days, the rats were anesthetized, the brains were excised, homogenized and used for the measurements of lipid peroxidation (LPO), thiol groups, activities of antioxidant enzymes, DNA damage and tumor necrosis factor-α (TNF-α), neuronal nitric oxide synthase (nNOS) and poly (ADP-ribose) polymerase-1 (PARP-1) expression. The results were analyzed using the Kruskal-Wallis and Conover-Inman tests or ANOVA and the Tukey-Kramer test. Doxorubicin, PTX and DTX induced OS, DNA damage and changes in expression of TNF-α, nNOS and PARP-1 in the rat brain. Pirolin alone increased LPO, manganese superoxide dismutase (MnSOD) and catalase (CAT) activities and the expression of PARP-1 but decreased TNF-α expression. PL, in combination with anticancer drugs, partially protected the rat brain against the toxic effects of DOX and taxanes. The best protective effects of PL were obtained with PTX. Pirolin partially attenuated brain damage caused by DOX/taxanes, highlighting its potential application in protecting the brain against DOX-, DTX- and PTX-evoked OS.


Subject(s)
Antineoplastic Agents/toxicity , Cyclic N-Oxides/pharmacology , Doxorubicin/toxicity , Neuroprotective Agents/pharmacology , Paclitaxel/toxicity , Taxoids/toxicity , Animals , Brain/drug effects , Brain/metabolism , Catalase/metabolism , Comet Assay , Docetaxel , Male , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress/drug effects , Poly (ADP-Ribose) Polymerase-1/metabolism , Rats, Wistar , Sulfhydryl Compounds/metabolism , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Tumor Necrosis Factor-alpha/metabolism
14.
Mater Sci Eng C Mater Biol Appl ; 74: 167-176, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28254282

ABSTRACT

Brain cancer, up-regulated with transferrin receptor led to concept of transferrin receptor targeted anticancer therapeutics. Docetaxel loaded d-α-tocopherol polyethylene glycol 1000 succinate conjugated chitosan (TPGS-chitosan) nanoparticles were prepared with or without transferrin decoration. In vitro experiments using C6 glioma cells showed that docetaxel loaded chitosan nanoparticles, non-targeted and transferrin receptor targeted TPGS-chitosan nanoparticles have enhanced the cellular uptake and cytotoxicity. The IC50 values of non-targeted and transferrin receptor targeted nanoparticles from cytotoxic assay were found to be 27 and 148 folds, respectively higher than Docel™. In vivo pharmacokinetic study showed 3.23 and 4.10 folds enhancement in relative bioavailability of docetaxel for non-targeted and transferrin receptor targeted nanoparticles, respectively than Docel™. The results have demonstrated that transferrin receptor targeted nanoparticles could enhance the cellular internalization and cytotoxicity of docetaxel via transferrin receptor with improved pharmacokinetics for clinical applications.


Subject(s)
Chitosan/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Taxoids/chemistry , alpha-Tocopherol/chemistry , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel , Drug Carriers/chemical synthesis , Female , Half-Life , Humans , Male , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Nanoparticles/toxicity , Particle Size , Rats , Receptors, Transferrin/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Taxoids/pharmacokinetics , Taxoids/toxicity
15.
Cell Physiol Biochem ; 41(2): 583-597, 2017.
Article in English | MEDLINE | ID: mdl-28214878

ABSTRACT

BACKGROUND/AIMS: Accumulating evidence revealed that microRNAs (miRNAs) have been demonstrated as critical molecules in tumor development and progression. MiR-26a, located in a fragile chromosomal region associated with various human cancer, has been reported to be involved in regulating various cellular process, such as proliferation, apoptosis and invasion through targeting multiple oncogene. Docetaxel-mediated chemotherapy has been applied in improving the survival and prognosis of patients with advanced lung adenocarcinoma (LAD). However, chemoresistance remains a major impediment to clinical application of this agent. It has been presented that decreased miR-26a expression lead to cisplatin resistance and promoted growth and migration in human lung cancer. Enhancer of zeste homolog 2 (EZH2) is the target of miR-26a. The present study aimed to investigate the function of miR-26a/EZH2 in the acquisition of malignant behaviors of LAD. METHODS: MiR-26a and EZH2 expression levels in the dcetaxel-insensitive groups (n = 19) and the docetaxel-sensitive groups (n = 18) were assessed by qRT-PCR. Colony formation assay, flow cytometric analysis, wound healing assay, cell transwell assays and western blotting were performed to assess the effects of miR-26a on proliferation, apoptosis and epithelial-to-mesenchymal (EMT) phenotypes in docetaxel resistant LAD cells in vitro. Xenograft transplantation, immunohistochemistry, tunel assays and western blotting assays were employed to demonstrate the role of miR-26a in docetaxel resistant LAD cells in vivo. The expression level of EZH2 in docetaxel-resistant LAD cells and corresponding parental cells was detected by qRT-PCR and western blotting. The relationship between miR-26a and EZH2 was confirmed by luciferase reporter assay. And rescue assays were performed to further confirm that miRNA-26a contributes to the acquisition of malignant behaviors of docetaxel-resistant LAD cells through targeting EZH2. RESULTS: MiR-26a was significantly down-regulated in the dcetaxel-insensitive groups (n = 19) compared with the docetaxel-sensitive groups (n = 18) assessed by qRT-PCR. MiR-26a decreased the proliferation, increased the apoptosis rate and reversed EMT to MET of docetaxel-resistant LAD cells both in vivo and vitro. EZH2 was confirmed as target of miR-26a. Rescue assays further verified that the function of miR-26a exerts in docetaxel-resistant LAD cells is through targeting EZH2. CONCLUSIONS: Our data revealed that overexpression of miR-26a in docetaxel-resistant LAD cells could decrease the proliferation, increase the apoptosis rate and reverse EMT to MET of docetaxel-resistant LAD cells both in vivo and vitro and such function is partially exerted via downregulating EZH2. MiR-26a/EZH2 signal pathway makes contribute to the malignant phenotype of docetaxel-resistant of LAD cells which indicated that miR-26a exerts pivotal functions in the molecular etiology of chemoresistant lung adenocarcinoma.


Subject(s)
Adenocarcinoma/pathology , Drug Resistance, Neoplasm , Enhancer of Zeste Homolog 2 Protein/metabolism , Lung Neoplasms/pathology , MicroRNAs/metabolism , 3' Untranslated Regions , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Antagomirs/metabolism , Apoptosis/drug effects , Base Sequence , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Docetaxel , Drug Resistance, Neoplasm/drug effects , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Epithelial-Mesenchymal Transition , Humans , Lung Neoplasms/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Neoplasm Staging , RNA Interference , RNA, Small Interfering/metabolism , Sequence Alignment , Taxoids/toxicity
16.
Eur J Pharm Biopharm ; 114: 186-193, 2017 May.
Article in English | MEDLINE | ID: mdl-28161551

ABSTRACT

BACKGROUND: Docetaxel is a cytostatic agent approved for treatment of non-small cell lung cancer as well as other cancers. Although docetaxel is an effective cytostatic agent, its effectiveness in clinical practice is associated with a variety of acute and long term side-effects. To overcome systemic side-effects, a slow release formulation based on calcium sulfate with docetaxel for intra-tumoral administration was developed. METHODS: Two formulations with the calcium sulfate NanoZolid technology were generated with a twofold difference in docetaxel drug load. The formulations were injected intra-tumorally as a paste which solidified within the tumor. The effects of the two intra-tumoral injection formulations were tested in female mice (n=60) inoculated with subcutaneous Lewis lung carcinoma cells. The two formulations were compared to systemic intraperitoneal injection of docetaxel and a placebo formulation without docetaxel. Tumor volumes were measured and systemic side-effects were evaluated using body weight and cell counts from whole blood as well as plasma concentrations. RESULTS: Both docetaxel formulations showed a significantly higher antitumor efficacy compared to placebo, which was comparable to that of systemic administration of docetaxel. Moreover, the intra-tumoral formulations with docetaxel showed reduced systemic toxicity compared to systemic treatment, including less weight loss and no decrease in blood cell counts. CONCLUSIONS: The results suggest that intra-tumoral slow release calcium sulfate based formulations with docetaxel can be an alternative strategy as an efficient local antitumoral treatment with reduced systemic toxicity.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Calcium Sulfate/chemistry , Taxoids/administration & dosage , Taxoids/pharmacology , Animals , Antineoplastic Agents, Phytogenic/toxicity , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/pathology , Docetaxel , Drug Compounding , Excipients , Female , Humans , Injections, Intralesional , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Taxoids/toxicity
17.
J Biomed Nanotechnol ; 13(4): 393-408, 2017 Apr.
Article in English | MEDLINE | ID: mdl-29384603

ABSTRACT

In this work, docetaxel (DTX) was encapsulated in monomethoxy poly(ethylene glycol)-poly(ε-caprolactone) (mPEG-PCL) micelles and monomethoxy poly(ethylene glycol)-poly(D, L-lactic acid) (mPEG-PLA) micelles, respectively. For the further application, the acute/genetic toxicity evaluation and pharmacokinetic/pharmacodynamic study of the two kinds of micellar nanomedicines were performed. In the study of anticancer activity in vitro and in vivo, DTX micelles showed better tumorgrowth inhibition than free DTX. The pharmacokinetic and tissue distribution studies showed that the DTX incorporated in micelles (especially in DTX-mPEG-PCL) retained significantly higher concentration in plasma and tumor tissue compared with free DTX. The acute toxicity and genotoxicity studies indicated that DTX micelles were safer than the docetaxel injection in cancer therapy and DTX-mPEG-PCL had less damage to DNA than DTX-mPEG-PLA. So the micelles had a pronounced effect on reducing acute toxicity and genotoxicity of docetaxel. In conclusion, DTX micelles were efficient and safe on breast carcinoma chemotherapy.


Subject(s)
Nanocapsules/administration & dosage , Nanocapsules/toxicity , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Taxoids/administration & dosage , Taxoids/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Docetaxel , Dose-Response Relationship, Drug , Humans , MCF-7 Cells , Metabolic Clearance Rate , Mice , Micelles , Nanocapsules/ultrastructure , Neoplasms, Experimental/pathology , Organ Specificity , Particle Size , Taxoids/toxicity , Tissue Distribution , Treatment Outcome
18.
Xenobiotica ; 47(4): 290-296, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27218144

ABSTRACT

1. Magnesium isoglycyrrhizinate (MgIg) has been extensively used in treating liver injury which is the common adverse reaction of docetaxel (DOC). Due to the narrow therapeutic window, small changes in pharmacokinetic profiles can alter the toxicity and therapeutic efficacy of DOC significantly. The study aimed to explore the effects of MgIg on the disposition of DOC and the potential mechanism in DOC-induced liver injury. 2. Pharmacokinetics and tissues distribution behaviors showed that there was no significant difference between DOC group (DOCG) and MgIg + DOC group (MDOCG). The mRNA and protein levels of cytochrome P450 3A1 (CYP3A1) in liver, intestine, and kidney were significantly upregulated, and the P-glycoprotein (P-gp) was obviously downregulated in MDOCG when compared with DOCG. 3. Immunoglobulin M (IgM), CD8+ were upregulated in DOCG; while in MDOCG, IgM, CD8+ recovered to normal levels and complement C3; CD4+ were upregulated. 4. MgIg had no significant effects on the disposition of DOC in docetaxel-induced liver injury. Additional, potential drug-drug interaction may happen if MgIg co-administered with antitumor drugs which are the substrates of CYP3A4 or P-gp. Hepatoprotective mechanism of MgIg perhaps was through upregulation of C3, CD4+ and downregulation of IgM, CD8+.


Subject(s)
Saponins/toxicity , Taxoids/toxicity , Triterpenes/toxicity , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Cytochrome P-450 Enzyme System/metabolism , Docetaxel , Drug Interactions , Humans , Saponins/chemistry , Taxoids/chemistry , Triterpenes/chemistry
19.
Brain Imaging Behav ; 11(6): 1652-1663, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27766586

ABSTRACT

Women with ovarian cancer often undergo chemotherapy involving multiple agents. However, little is known about treatment-related central neurotoxicity in this population. The goal of this cross-sectional study was to assess brain structure and function and neurocognitive abilities in patients with ovarian cancer following first-line chemotherapy. Eighteen patients with ovarian, peritoneal and fallopian tube cancer and eighteen healthy controls matched for gender, age and education participated in the study. The patients were evaluated 1-4 months following completion of first-line taxane/platinum chemotherapy. All participants underwent structural and functional magnetic resonance imaging (MRI), and completed neuropsychological tests of attention, memory and executive functions. Neuroimaging assessments included voxel-based morphometry (VBM) for measuring gray matter (GM) volume, and functional MRI (fMRI) during the N-back working memory task. The results of VBM showed that patients had significantly reduced GM volume compared to healthy controls in the right middle/superior frontal gyrus, and in the left supramarginal gyrus and left inferior parietal lobule. fMRI results indicated significantly decreased activation in patients relative to healthy controls in the left middle frontal gyrus and left inferior parietal lobule during the N-back task (1/2/3-back >0-back). There were no statistically significant differences between the two groups on the neuropsychological tests. This is the first study showing structural and functional alterations involving frontal and parietal regions in patients with ovarian cancer treated with first-line chemotherapy. These findings are congruent with studies involving women with breast cancer, and provide additional supporting evidence for central neurotoxicity associated with taxane/platinum chemotherapy.


Subject(s)
Antineoplastic Agents/toxicity , Brain/diagnostic imaging , Brain/physiopathology , Ovarian Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents/therapeutic use , Brain/drug effects , Brain/pathology , Brain Mapping , Bridged-Ring Compounds/therapeutic use , Bridged-Ring Compounds/toxicity , Cohort Studies , Cross-Sectional Studies , Female , Gray Matter/diagnostic imaging , Gray Matter/drug effects , Gray Matter/pathology , Gray Matter/physiopathology , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Memory, Short-Term/physiology , Middle Aged , Neuropsychological Tests , Organ Size , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/physiopathology , Ovarian Neoplasms/psychology , Pilot Projects , Platinum Compounds/therapeutic use , Platinum Compounds/toxicity , Preliminary Data , Taxoids/therapeutic use , Taxoids/toxicity
20.
Sci Rep ; 6: 38787, 2016 12 09.
Article in English | MEDLINE | ID: mdl-27934917

ABSTRACT

Palmitoyl ascorbate (PA) as an antioxidant has the potential for the treatment of cancer. In the present study, a nanocarrier system was developed for co-delivery of docetaxel (DOC) with palmitoyl ascorbate and the therapeutic efficacy of a combination drug regimen was investigated. For this purpose, different ratios of docetaxel and palmitoyl ascorbate were co-encapsulated in a liposome and they all showed high encapsulation efficiency. The average diameters of the liposomes ranged from 140 to 170 nm. Negative zeta potential values were observed for all systems, ranged from -40 mV to -56 mV. Studies on drug release and cellular uptake of the co-delivery system demonstrated that both drugs were effectively taken up by the cells and released slowly. Moreover, the liposome loading drugs with DOC/PA concentration ratio of 1:200 showed the highest anti-tumor activity to three different types of tumor cells. The higher in vivo therapeutic efficacy with lower systemic toxicity of the DOC-PA200-LPs was also verified by the H22 tumor bearing mice model. Our results showed that such co-loaded delivery systems could serve as a promising therapeutic approach to improve clinical outcomes against hepatic carcinoma.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Antioxidants/administration & dosage , Ascorbic Acid/analogs & derivatives , Taxoids/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Agents, Phytogenic/toxicity , Antioxidants/therapeutic use , Antioxidants/toxicity , Ascorbic Acid/administration & dosage , Ascorbic Acid/therapeutic use , Ascorbic Acid/toxicity , Cell Line, Tumor , Docetaxel , Drug Compounding , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Liposomes/administration & dosage , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Mice, Inbred ICR , Nanocapsules , Particle Size , Pharmaceutical Vehicles , Taxoids/toxicity , Tumor Burden
SELECTION OF CITATIONS
SEARCH DETAIL
...