Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
PLoS One ; 16(11): e0258605, 2021.
Article in English | MEDLINE | ID: mdl-34780478

ABSTRACT

BACKGROUND: The phase III KEYNOTE-604 study confirmed the benefit of pembrolizumab combined with chemotherapy in the first-line treatment of extensive-stage small-cell lung cancer (ES-SCLC). Taken into account the clinical benefits of pembrolizumab and its high cost, this study aimed to assess the cost-effectiveness of adding pembrolizumab to standard first-line etoposide-platinum (EP) for patients with ES-SCLC from the US payer perspective. METHODS: A Markov model was developed to compare the cost and quality-adjusted life-year (QALY) of pembrolizumab plus EP and placebo plus EP over a 10-year time horizon. Clinical efficacy and safety data were pooled from the KEYNOTE-604 trial. Utilities were obtained from published resources. Costs were mainly collected from Medicare in 2020. Sensitivity analyses were performed to examine the robustness of our model. RESULTS: Adding pembrolizumab to standard first-line EP resulted in the better effectiveness than EP chemotherapy alone for ES-SCLC by 0.22 QALYs. Pembrolizumab plus EP was dominated economically by placebo plus EP, leading to an incremental cost-effectiveness ratio (ICER) of $334,373/ QALY. Deterministic sensitivity analyses indicated that the uncertainty in model parameters exerted no substantial effect on our results. Probability sensitivity analysis indicated that probabilities for pembrolizumab plus EP being cost-effective within a wide range of willingness to pay were modest. CONCLUSION: From the US payer perspective, the first-line treatment for ES-SCLC with pembrolizumab plus EP was not cost-effective compared with placebo plus EP. Although pembrolizumab combination chemotherapy was beneficial to the survival of ES-SCLC, price reduction may be the necessary to improve its cost-effectiveness.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cost-Benefit Analysis/methods , Etoposide/therapeutic use , Immunotherapy/methods , Lung Neoplasms/drug therapy , Platinum/therapeutic use , Small Cell Lung Carcinoma/drug therapy , Antibodies, Monoclonal, Humanized/economics , Antineoplastic Agents, Immunological/economics , Antineoplastic Agents, Phytogenic/economics , Antineoplastic Combined Chemotherapy Protocols/economics , Double-Blind Method , Etoposide/economics , Follow-Up Studies , Humans , Lung Neoplasms/epidemiology , Lung Neoplasms/pathology , Markov Chains , Medicare , Neoplasm Staging , Progression-Free Survival , Quality-Adjusted Life Years , Small Cell Lung Carcinoma/epidemiology , Small Cell Lung Carcinoma/pathology , United States/epidemiology
2.
JAMA Netw Open ; 3(11): e2027074, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33226431

ABSTRACT

Importance: The neoadjuvant treatment options for ERBB2-positive (also known as HER2-positive) breast cancer are associated with different rates of pathologic complete response (pCR). The KATHERINE trial showed that adjuvant trastuzumab emtansine (T-DM1) can reduce recurrence in patients with residual disease compared with patients treated with trastuzumab; however, T-DM1 and other ERBB2-targeted agents are costly, and understanding the costs and health consequences of various combinations of neoadjuvant followed by adjuvant treatments in the United States is needed. Objective: To examine the costs and disease outcomes associated with selection of various neoadjuvant followed by adjuvant treatment strategies for patients with ERBB2-positive breast cancer. Design, Setting, and Participants: In this economic evaluation, a decision-analytic model was developed to evaluate various neoadjuvant followed by adjuvant treatment strategies for women with ERBB2-positive breast cancer from a health care payer perspective in the United States. The model was informed by the KATHERINE trial, other clinical trials with different regimens from the KATHERINE trial, the Flatiron Health Database, McKesson Corporation data, and other evidence in the published literature. Starting trial median age for KATHERINE patients was 49 years (range, 24-79 years in T-DM1 arm and 23-80 years in trastuzumab arm). The model simulated patients receiving 5 different neoadjuvant followed by adjuvant treatment strategies. Data analyses were performed from March 2019 to August 2020. Exposure: There were 4 neoadjuvant regimens: (1) HP: trastuzumab (H) plus pertuzumab (P), (2) THP: paclitaxel (T) plus H plus P, (3) DDAC-THP: dose-dense anthracycline/cyclophosphamide (DDAC) plus THP, (4) TCHP: docetaxel (T) plus carboplatin (C) plus HP. All patients with pCR, regardless of neoadjuvant regimen, received adjuvant H. Patients with residual disease received different adjuvant therapies depending on the neoadjuvant regimen according to the 5 following strategies: (1) neoadjuvant DDAC-THP followed by adjuvant H, (2) neoadjuvant DDAC-THP followed by adjuvant T-DM1, (3) neoadjuvant THP followed by adjuvant DDAC plus T-DM1, (4) neoadjuvant HP followed by adjuvant DDAC/THP plus T-DM1, or (5) neoadjuvant TCHP followed by adjuvant T-DM1. Main Outcomes and Measures: Lifetime costs in 2020 US dollars and quality-adjusted life-years (QALYs) were estimated for each treatment strategy, and incremental cost-effectiveness ratios were estimated. A strategy was classified as dominated if it was associated with fewer QALYs at higher costs than the alternative. Results: In the base-case analysis, costs ranged from $415 833 (strategy 3) to $518 859 (strategy 4), and QALYs ranged from 9.67 (strategy 1) to 10.73 (strategy 3). Strategy 3 was associated with the highest health benefits (10.73 QALYs) and lowest costs ($415 833) and dominated all other strategies. Probabilistic analysis confirmed that this strategy had the highest probability of cost-effectiveness (>70% at willingness-to-pay thresholds of $0-200,000/QALY) and was associated with the highest net benefit. Conclusions and Relevance: These results suggest that neoadjuvant THP followed by adjuvant H for patients with pCR or followed by adjuvant DDAC plus T-DM1 for patients with residual disease was associated with the highest health benefits and lowest costs for women with ERBB2-positive breast cancer compared with other treatment strategies considered.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/therapy , Neoadjuvant Therapy/economics , Receptor, ErbB-2/genetics , Ado-Trastuzumab Emtansine/economics , Ado-Trastuzumab Emtansine/therapeutic use , Adult , Aged , Anthracyclines/economics , Anthracyclines/therapeutic use , Antibodies, Monoclonal, Humanized/economics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/economics , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Phytogenic/economics , Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/pathology , Case-Control Studies , Cost-Benefit Analysis , Cross-Linking Reagents/economics , Cross-Linking Reagents/therapeutic use , Drug Therapy, Combination , Female , Humans , Immunosuppressive Agents/economics , Immunosuppressive Agents/therapeutic use , Middle Aged , Paclitaxel/economics , Paclitaxel/therapeutic use , Quality-Adjusted Life Years , Trastuzumab/economics , Trastuzumab/therapeutic use , Tubulin Modulators/economics , Tubulin Modulators/therapeutic use , United States/epidemiology
3.
Biochim Biophys Acta Rev Cancer ; 1874(1): 188381, 2020 08.
Article in English | MEDLINE | ID: mdl-32492470

ABSTRACT

The United States Food and Drug Administration has permitted number of therapeutic agents for cancer treatment. Most of them are expensive and have some degree of systemic toxicity which makes overbearing in clinical settings. Although advanced research continuously applied in cancer therapeutics, but drug resistance, metastasis, and recurrence remain unanswerable. These accounts to an urgent clinical need to discover natural compounds with precisely safe and highly efficient for the cancer prevention and cancer therapy. Gambogic acid (GA) is the principle bioactive and caged xanthone component, a brownish gamboge resin secreted from the of Garcinia hanburyi tree. This molecule showed a spectrum of biological and clinical benefits against various cancers. In this review, we document distinct biological characteristics of GA as a novel anti-cancer agent. This review also delineates specific molecular mechanism(s) of GA that are involved in anti-cancer, anti-metastasis, anti-angiogenesis, and chemo-/radiation sensitizer activities. Furthermore, recent evidence, development, and implementation of various nanoformulations of gambogic acid (nanomedicine) have been described.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Nanomedicine/methods , Neoplasms/therapy , Radiation-Sensitizing Agents/administration & dosage , Xanthones/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/economics , Chemoradiotherapy/economics , Chemoradiotherapy/methods , Clinical Trials, Phase II as Topic , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Costs , Garcinia/chemistry , Humans , Nanomedicine/economics , Nanoparticles/chemistry , Neoplasms/economics , Radiation-Sensitizing Agents/economics , Resins, Plant/chemistry , Treatment Outcome , Xanthones/economics , Xenograft Model Antitumor Assays
4.
J Appl Microbiol ; 129(2): 345-355, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32091657

ABSTRACT

AIMS: Paclitaxel is a type of broad-spectrum anticancer drug in short supply. The price of acetyl-CoA (17 709 677·4 USD mol-1 ), which is the acetyl group donor for the enzymatic synthesis of the intermediate, baccatin Ⅲ, is still the bottleneck of the mass production of paclitaxel. This study reports a novel acetyl group donor, which could substantially reduce the cost of production. METHODS AND RESULTS: In this study, a substrate spectrum with 14 kinds of representative acetyl-donor substitutes predicted by computer-aided methods was tested in a 10-deacetylbaccatin Ⅲ-10-O-acetyltransferase (DBAT) heterogeneous-expressed open-whole-cell catalytic system. The results of computer prediction and experimental analysis revealed the rule of the acetyl-donor compounds based on this substrate spectrum. N-acetyl-d-glucosamine (30·95 USD mol-1 , about 572 202-fold cheaper than acetyl-CoA) is selected as a suitable substitute under the rule. The yield when using N-acetyl-d-glucosamine as acetyl donor in open-whole-cell catalytic system was 2·13-fold of that when using acetyl-CoA. In the in vivo system, the yield increased 24·17%, which may indicate its cooperation with acetyl-CoA. CONCLUSION: The success of open-whole-cell synthesis and in vivo synthesis of baccatin Ⅲ by adding N-acetyl-d-glucosamine as acetyl substrate demonstrates that it is a useful substrate to improve the yield of baccatin Ⅲ. SIGNIFICANCE AND IMPACT OF THE STUDY: All these findings provided a potential acetyl-donor substitute for acetyl-CoA, as well as a low cost and efficient method of preparing paclitaxel through baccatin Ⅲ semi-synthesis.


Subject(s)
Acetylglucosamine/metabolism , Alkaloids/biosynthesis , Acetyl Coenzyme A/metabolism , Acetyltransferases/genetics , Acetyltransferases/metabolism , Alkaloids/economics , Antineoplastic Agents, Phytogenic/biosynthesis , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/economics , Biocatalysis , Paclitaxel/biosynthesis , Paclitaxel/chemistry , Paclitaxel/economics , Substrate Specificity , Taxoids/economics
5.
J Invasive Cardiol ; 32(1): 18-24, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31611426

ABSTRACT

BACKGROUND: A meta-analysis of trials in endovascular therapy suggested an increased mortality associated with treatment exposure to paclitaxel. Multiple publications and corrections of prior data were performed, and the United States Food and Drug Administration has issued multiple advisories regarding paclitaxel use. We analyzed how this controversy impacted device purchasing and related utilization patterns in the period immediately following publication of the meta-analysis. METHODS AND RESULTS: Ascension Healthcare System purchase data over a 14-month period were synthesized across centers for both paclitaxel and non-paclitaxel devices. A fixed-effects regression model and a binary regression model with facility-level controls were used to compare purchasing patterns before and after the meta-analysis. Purchase volumes of each paclitaxel device fell. Pooled purchase volumes of all paclitaxel devices decreased from a 14-month peak of 631 devices in October 2018 to a 14-month nadir of 359 devices in February 2019. An F-test comparing the pooled-month specific fixed effects for the months before vs after the publication of the meta-analysis has an F-statistic of 11.64, suggesting that average purchasing levels in the two periods are statistically different (P<.001). Utilization of non-paclitaxel devices did not decline. CONCLUSIONS: Purchase volumes of paclitaxel devices decreased immediately during the months following publication of the related meta-analysis. Total Ascension-wide paclitaxel device purchase volume in February 2019 demonstrated a 43.1% reduction from peak monthly purchase volume during the assessed period and a 32.5% reduction compared with November 2019, the last month preceding publication of the meta-analysis.


Subject(s)
Drug-Eluting Stents , Endovascular Procedures , Graft Occlusion, Vascular , Long Term Adverse Effects , Paclitaxel , Peripheral Arterial Disease/surgery , Product Surveillance, Postmarketing , Antineoplastic Agents, Phytogenic/economics , Antineoplastic Agents, Phytogenic/pharmacology , Coated Materials, Biocompatible/pharmacology , Consumer Product Safety , Drug-Eluting Stents/adverse effects , Drug-Eluting Stents/economics , Drug-Eluting Stents/statistics & numerical data , Endovascular Procedures/adverse effects , Endovascular Procedures/instrumentation , Graft Occlusion, Vascular/diagnosis , Graft Occlusion, Vascular/mortality , Humans , Long Term Adverse Effects/etiology , Long Term Adverse Effects/mortality , Meta-Analysis as Topic , Neointima/prevention & control , Paclitaxel/economics , Paclitaxel/pharmacology , Product Surveillance, Postmarketing/economics , Product Surveillance, Postmarketing/methods
6.
Mol Biotechnol ; 60(7): 492-505, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29796788

ABSTRACT

Natural production of anti-cancer drug taxol from Taxus has proved to be environmentally unsustainable and economically unfeasible. Currently, bioengineering the biosynthetic pathway of taxol is an attractive alternative production approach. 10-deacetylbaccatin III-10-O-acetyl transferase (DBAT) was previously characterized as an acyltransferase, using 10-deacetylbaccatin III (10-DAB) and acetyl CoA as natural substrates, to form baccatin III in the taxol biosynthesis. Here, we report that other than the natural acetyl CoA (Ac-CoA) substrate, DBAT can also utilize vinyl acetate (VA), which is commercially available at very low cost, acylate quickly and irreversibly, as acetyl donor in the acyl transfer reaction to produce baccatin III. Furthermore, mutants were prepared via a semi-rational design in this work. A double mutant, I43S/D390R was constructed to combine the positive effects of the different single mutations on catalytic activity, and its catalytic efficiency towards 10-DAB and VA was successfully improved by 3.30-fold, compared to that of wild-type DBAT, while 2.99-fold higher than the catalytic efficiency of WT DBAT towards 10-DAB and Ac-CoA. These findings can provide a promising economically and environmentally friendly method for exploring novel acyl donors to engineer natural product pathways.


Subject(s)
Acetyltransferases/genetics , Alkaloids/biosynthesis , Antineoplastic Agents, Phytogenic/biosynthesis , Taxus/enzymology , Acetyltransferases/chemistry , Acetyltransferases/metabolism , Alkaloids/economics , Antineoplastic Agents, Phytogenic/economics , Bioengineering , Biosynthetic Pathways , Computational Biology , Cost-Benefit Analysis , Genetic Engineering , Models, Molecular , Mutagenesis , Paclitaxel/biosynthesis , Paclitaxel/economics , Substrate Specificity , Taxoids/economics , Taxoids/metabolism , Taxus/chemistry , Taxus/genetics , Taxus/metabolism , Vinyl Compounds/chemistry , Vinyl Compounds/metabolism
7.
Food Funct ; 9(4): 1978-1992, 2018 Apr 25.
Article in English | MEDLINE | ID: mdl-29594287

ABSTRACT

Citrus medica (Citron) is an underutilized fruit plant having various bioactive components in all parts of the plant. The major bioactive compounds present are iso-limonene, citral, limonene, phenolics, flavonones, vitamin C, pectin, linalool, decanal, and nonanal, accounting for several health benefits. Pectin and heteropolysachharides also play a major role as dietary fibers. The potential impact of citron and its bioactive components to prevent or reverse destructive deregulated processes responsible for certain diseases has attracted different researchers' attention. The fruit has numerous nutraceutical benefits, proven by pharmacological studies; for example, anti-catarrhal, capillary protector, anti-hypertensive, diuretic, antibacterial, antifungal, anthelmintic, antimicrobial, analgesic, strong antioxidant, anticancerous, antidiabetic, estrogenic, antiulcer, cardioprotective, and antihyperglycemic. The present review explores new insights into the benefits of citron in various body parts. Throughout the world, citron has been used in making carbonated drinks, alcoholic beverages, syrup, candied peels, jams, marmalade, cordials, and many other value added products, which suggests it is an appropriate raw material to develop healthy processed food. In the present review, the fruit taxonomical classification, beneficial phytochemicals, antioxidant activities, and health benefits are discussed.


Subject(s)
Chronic Disease/prevention & control , Citrus , Fruit , Functional Food , Animals , Antineoplastic Agents, Phytogenic/analysis , Antineoplastic Agents, Phytogenic/economics , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/therapeutic use , Antioxidants/analysis , Antioxidants/economics , Antioxidants/isolation & purification , Antioxidants/therapeutic use , Citrus/chemistry , Citrus/economics , Dietary Supplements/analysis , Dietary Supplements/economics , Food Additives/chemistry , Food Additives/economics , Food Additives/isolation & purification , Food Additives/therapeutic use , Food-Processing Industry/economics , Fruit/chemistry , Fruit/economics , Humans , Industrial Waste/analysis , Industrial Waste/economics , Nutritive Value , Phytochemicals/analysis , Phytochemicals/economics , Phytochemicals/isolation & purification , Phytochemicals/therapeutic use , Plant Extracts/chemistry , Plant Extracts/economics , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use
8.
Pharmacoeconomics ; 36(10): 1153-1163, 2018 10.
Article in English | MEDLINE | ID: mdl-29600384

ABSTRACT

As part of the single technology appraisal (STA) process, the National Institute for Health and Care Excellence (NICE) invited Celgene Ltd to submit clinical and cost-effectiveness evidence for paclitaxel as albumin-bound nanoparticles (Nab-Pac) in combination with gemcitabine (Nab-Pac + Gem) for patients with untreated metastatic pancreatic cancer. The STA was a review of NICE's 2015 guidance (TA360) in which Nab-Pac + Gem was not recommended for patients with untreated metastatic pancreatic cancer. The review was prompted by a proposed Patient Access Scheme (PAS) discount on the price of Nab-Pac and new evidence that might lead to a change in the guidance. The Liverpool Reviews and Implementation Group at the University of Liverpool was the Evidence Review Group (ERG). This article summarises the ERG's review of the company's evidence submission for Nab-Pac + Gem, and the Appraisal Committee (AC) decision. The final scope issued by NICE listed three comparators: gemcitabine monotherapy (Gem), gemcitabine in combination with capecitabine (Gem + Cap), and a combination of oxaliplatin, irinotecan, leucovorin and fluorouracil (FOLFIRINOX). Clinical evidence for the comparison of Nab-Pac + Gem versus Gem was from the phase III CA046 randomized controlled trial. Analysis of progression-free survival (PFS) and overall survival (OS) showed statistically significant improvement for patients treated with Nab-Pac + Gem versus Gem. Clinical evidence for the comparison of Nab-Pac + Gem versus FOLFIRINOX and versus Gem + Cap was derived from a network meta-analysis (NMA). Results of the NMA did not indicate a statistically significant difference in OS or PFS for the comparison of Nab-Pac + Gem versus either Gem + Cap or FOLFIRINOX. The ERG's main concerns with the clinical effectiveness evidence were difficulties in identifying the patient population for whom treatment with Nab-Pac + Gem is most appropriate, and violation of the proportional hazards (PH) assumption in the CA046 trial. The ERG highlighted methodological issues in the cost-effectiveness analysis pertaining to the modelling of survival outcomes, estimation of drug costs and double counting of adverse-event disutilities. The AC accepted all the ERG's amendments to the company's cost-effectiveness model; however, these did not make important differences to the incremental cost-effectiveness ratios (ICERs). The company's base-case ICER was £46,932 per quality-adjusted life-year (QALY) gained for the comparison of Nab-Pac + Gem versus Gem. Treatment with Nab-Pac + Gem was dominated both by treatment with Gem + Cap and with FOLFIRINOX in the company's base case. The AC concluded that the most plausible ICER for treatment with Nab-Pac + Gem versus Gem was in the range of £41,000-£46,000 per QALY gained. The AC concluded that Nab-Pac + Gem was not cost effective compared with Gem + Cap or FOLFIRINOX, and accepted that treatment with Nab-Pac + Gem met the end-of-life criteria versus Gem but did not consider Nab-Pac + Gem to meet the end-of-life criteria compared with Gem + Cap or FOLFIRINOX. The AC also concluded that although patients who would receive Nab-Pac + Gem rather than FOLFIRINOX or Gem + Cap were difficult to distinguish, they were identifiable in clinical practice. The AC recommended treatment with Nab-Pac + Gem for patients with untreated metastatic pancreatic cancer for whom other combination chemotherapies were unsuitable and who would otherwise receive Gem.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/economics , Cost-Benefit Analysis/statistics & numerical data , Deoxycytidine/analogs & derivatives , Paclitaxel/economics , Pancreatic Neoplasms/economics , Technology Assessment, Biomedical/statistics & numerical data , Antimetabolites, Antineoplastic/economics , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents, Phytogenic/economics , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Capecitabine/economics , Capecitabine/therapeutic use , Deoxycytidine/economics , Deoxycytidine/therapeutic use , Disease-Free Survival , Fluorouracil/economics , Fluorouracil/therapeutic use , Humans , Irinotecan/economics , Irinotecan/therapeutic use , Leucovorin/economics , Leucovorin/therapeutic use , Models, Economic , Nanoparticles/economics , Nanoparticles/therapeutic use , Oxaliplatin/economics , Oxaliplatin/therapeutic use , Paclitaxel/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/secondary , Gemcitabine
9.
Farm Hosp ; 41(4): 550-558, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28683707

ABSTRACT

Prostate cancer (PC) is the most common urogenital malignancy in older men and the second leading cause of death by cancer in men in Europe. Current therapeutic practice considers Androgen Deprivation Therapy (ADT) as first line treatment for clinically localized prostate cancer at high-risk, either locally advanced or metastatic. ADT can be achieved through orchiectomy (surgical castration), luteinizing hormone-releasing hormone (LHRH) agonists, or through complete androgen blockade (LHRH agonist combined with an anti-androgen). Docetaxel in combination with prednisone or prednisolone is indicated for the treatment of patients with hormone-refractory metastatic prostate cancer. The CHAARTED and STAMPEDE clinical trials studied the effect of bringing forward the use of docetaxel added on to ADT in the context of hormone-sensitive patients. The CHAARTED clinical trial showed a significant increase in a variable with maximum relevance such as Overall Survival (OS), with a difference of 13.6 months between medians. There was also clinical benefit in the secondary variables: median time until castration-resistant disease or until clinical progression. In the STAMPEDE clinical trial, which included 39% of non-metastatic patients, a 10-month difference between medians was demonstrated in OS, and 17 months in the primary co-variable of Progression Free Survival. The most frequent adverse events were: neutropenia, febrile neutropenia, leucopenia, and general disorders such as asthenia, lethargy or fever. According to data from the CHAARTED and STAMPEDE studies, and the incremental cost of € 3 196.98 for adding on docetaxel to standard treatment, the estimated additional cost for each year of life gained is compatible with an incremental cost-effectiveness ratio between € 2 267.36 and € 3 851.78. In view of the efficacy and safety results, the proposed positioning is: to advance the use of docetaxel added to androgen deprivation therapy to first-line metastatic hormone-sensitive prostate cancer, regardless of metastatic volume, in those patients who meet the CHAARTED study criteria.


El cáncer de próstata (CP) es el tumor maligno urogenital más frecuente en hombres de edad avanzada y la segunda causa de muerte por cáncer en hombres en Europa. La práctica terapéutica actual considera como primera línea la terapia de privación de andrógenos (ADT; androgen deprivation therapy) en el cáncer de próstata clínicamente localizado de alto riesgo, localmente avanzado o metástasico. La ADT se realiza mediante orquiectomía (castración quirúrgica), agonistas de la hormona liberadora de hormona luteinizante (LHRH), o bien mediante el bloqueo androgénico completo (agonista LHRH más antiandrógeno). El docetaxel en combinación con prednisona o prednisolona está indicado para el tratamiento de pacientes con cáncer de próstata hormono-refractariometastásico. Los ensayos clínicos CHAARTED y STAMPEDE analizaron el efecto de adelantar el uso de docetaxel añadido a la ADT en el contexto del paciente hormonosensible. En el ensayo CHAARTED se demostró un aumento significativo en la variable de máxima relevancia como es la supervivencia global (SG), con una diferencia de medianas de 13,6 meses. También se obtuvo beneficio clínico en las variables secundarias mediana de tiempo hasta enfermedad resistente a la castración o hasta progresión clínica. En el ensayo STAMPEDE, en el que se incluyeron un 39% de pacientes no metastásicos, se demostró una diferencia de medianas de 10 meses en la SG y de 17 meses en la covariable principal de supervivencia libre de progresión. Los eventos adversos más frecuentes fueron: neutropenia, neutropenia febril, leucopenia y alteraciones generales tales como astenia, letargia o fiebre. Según los datos del estudio CHAARTED y STAMPEDE y el coste incremental de 3.196,98 € de añadir docetaxel a la terapia de referencia, por cada año de vida ganado el coste adicional estimado es compatible con un coste/eficacia incremental (CEI) entre 2.267,36 € y 3.851,78 €. A la vista de los importantes resultados de eficacia y el perfil manejable de seguridad, el posicionamiento propuesto es adelantar el uso de docetaxel añadido a la ADT a la primera línea del cáncer de próstata metastásico hormonosensible, en aquellos pacientes que cumplan los criterios básicos del estudio CHAARTED.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Prostatic Neoplasms/drug therapy , Taxoids/therapeutic use , Androgen Antagonists/adverse effects , Androgen Antagonists/economics , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/economics , Clinical Trials as Topic , Cost-Benefit Analysis , Disease Progression , Docetaxel , Humans , Male , Prostatic Neoplasms/economics , Taxoids/adverse effects , Taxoids/economics , Treatment Outcome
10.
J Manag Care Spec Pharm ; 23(4): 416-426, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28345444

ABSTRACT

BACKGROUND: With the approval of several new treatments for metastatic castration-resistant prostate cancer (mCRPC), budgetary impact is a concern for health plan decision makers. Budget impact models (BIMs) are becoming a requirement in many countries as part of formulary approval or reimbursement decisions. Cabazitaxel is a second-generation taxane developed to overcome resistance to docetaxel and is approved for the treatment of patients with mCRPC previously treated with a docetaxel-containing regimen. OBJECTIVE: To estimate a 1-year projected budget impact of varying utilization rates of cabazitaxel as a second-line treatment for mCRPC following docetaxel, using a hypothetical U.S. private managed care plan with 1 million members. METHODS: A BIM was developed to evaluate costs for currently available treatment options for patients with mCRPC previously treated with docetaxel. Treatments included in the model were cabazitaxel, abiraterone acetate, enzalutamide, and radium-223, with utilization rates derived from market research data. Medication costs were calculated according to published pricing benchmarks factored by dosing and duration of therapy as stated in the prescribing information for each agent. Published rates and costs of grade 3-4 adverse events were also factored into the model. In addition, the model reports budget impact under 2 scenarios. In the first base-case scenario, patient out-of-pocket costs were subtracted from the total cost of treatment. In the second scenario, all treatment costs were assumed to be paid by the plan. RESULTS: In a hypothetical 1 million-member health plan population, 100 patients were estimated to receive second-line treatment for mCRPC after treatment with docetaxel. Using current utilization rates for the 4 agents of interest, the base-case scenario estimated the cost of second-line treatment after docetaxel to be $6,331,704, or $0.528 per member per month (PMPM). In a scenario where cabazitaxel use increases from the base-rate case of 24% to a hypothetical rate of 33%, the PMPM cost would decrease to $0.524, reflecting a cost saving of $0.004 PMPM and equating to incremental savings of $49,546, or $497 per patient per year (PPPY). In the second scenario, when out-of-pocket costs were not considered, the cost of second-line treatment after docetaxel was estimated as $6,733,594, or $0.561 PMPM. With a hypothetical increase in cabazitaxel use (24%-33%), the PMPM cost would decrease to $0.554, reflecting savings of $0.007 PMPM and equating to incremental savings of $86,136, or $864 PPPY. The primary driver of cost savings with increased cabazitaxel use was lower acquisition cost. One-way sensitivity analyses revealed that the model results were robust over a wide range of input values (utilization, prevalence, and population parameters). CONCLUSIONS: In the presented BIM, an increase in cabazitaxel use is expected to result in modest cost savings to the health plan. Patient coinsurance savings may also be realized based on applicable Medicare Part B and Part D calculations. This BIM presents an objective, comprehensive, robust, and user-adaptable tool that health plans and medical decision makers may use to evaluate potential economic impact of formulary and reimbursement decisions. DISCLOSURES: Research and analysis were funded by Sanofi US. The sponsor had the opportunity to review the final draft; however, the authors were responsible for all content and editorial decisions. Flannery, Drea, Hudspeth, and Miao are employees of Sanofi. Miao is an owner of stock in Sanofi. Corman, Gao, and Xue are employees of Pharmerit International and served as consultants to Sanofi during this study. All authors contributed to study design and data collection and analysis. The manuscript was written by Flannery, along with the other authors, and revised by all the authors.


Subject(s)
Antineoplastic Agents, Phytogenic/economics , Prostatic Neoplasms, Castration-Resistant/economics , Taxoids/economics , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/therapeutic use , Budgets , Computer Simulation , Cost Savings , Docetaxel , Drug Costs , Drug-Related Side Effects and Adverse Reactions/economics , Health Care Costs , Humans , Male , Managed Care Programs/economics , Models, Economic , Prostatic Neoplasms, Castration-Resistant/drug therapy , Taxoids/adverse effects , Taxoids/therapeutic use
11.
J Manag Care Spec Pharm ; 23(1): 64-73, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28025930

ABSTRACT

BACKGROUND: Treatment patterns for metastatic colorectal cancer (mCRC) patients have changed considerably over the last decade with the introduction of new chemotherapies and targeted biologics. These treatments are often administered in various sequences with limited evidence regarding their cost-effectiveness. OBJECTIVE: To conduct a pharmacoeconomic evaluation of commonly administered treatment sequences among elderly mCRC patients. METHODS: A probabilistic discrete event simulation model assuming Weibull distribution was developed to evaluate the cost-effectiveness of the following common treatment sequences: (a) first-line oxaliplatin/irinotecan followed by second-line oxaliplatin/irinotecan + bevacizumab (OI-OIB); (b) first-line oxaliplatin/irinotecan + bevacizumab followed by second-line oxaliplatin/irinotecan + bevacizumab (OIB-OIB); (c) OI-OIB followed by a third-line targeted biologic (OI-OIB-TB); and (d) OIB-OIB followed by a third-line targeted biologic (OIB-OIB-TB). Input parameters for the model were primarily obtained from the Surveillance, Epidemiology, and End Results-Medicare linked dataset for incident mCRC patients aged 65 years and older diagnosed from January 2004 through December 2009. A probabilistic sensitivity analysis was performed to account for parameter uncertainty. Costs (2014 U.S. dollars) and effectiveness were discounted at an annual rate of 3%. RESULTS: In the base case analyses, at the willingness-to-pay (WTP) threshold of $100,000/quality-adjusted life-year (QALY) gained, the treatment sequence OIB-OIB (vs. OI-OIB) was not cost-effective with an incremental cost-effectiveness ratio (ICER) per patient of $119,007/QALY; OI-OIB-TB (vs. OIB-OIB) was dominated; and OIB-OIB-TB (vs. OIB-OIB) was not cost-effective with an ICER of $405,857/QALY. Results similar to the base case analysis were obtained assuming log-normal distribution. Cost-effectiveness acceptability curves derived from a probabilistic sensitivity analysis showed that at a WTP of $100,000/QALY gained, sequence OI-OIB was 34% cost-effective, followed by OIB-OIB (31%), OI-OIB-TB (20%), and OIB-OIB-TB (15%). CONCLUSIONS: Overall, survival increases marginally with the addition of targeted biologics, such as bevacizumab, at first line and third line at substantial costs. Treatment sequences with bevacizumab at first line and targeted biologics at third line may not be cost-effective at the commonly used threshold of $100,000/QALY gained, but a marginal decrease in the cost of bevacizumab may make treatment sequences with first-line bevacizumab cost-effective. Future economic evaluations should validate the study results using parameters from ongoing clinical trials. DISCLOSURES: This study was supported in part by a grant from the Agency for Healthcare Research and Quality (R01-HS018956) and in part by a grant from the Cancer Prevention and Research Institute of Texas (RP130051), which were obtained by Du. The authors report no conflicts of interest. Study concept and design were primarily contributed by Parikh, along with the other authors. All authors participated in data collection, and Parikh took the lead in data interpretation and analysis, along with Lairson and Morgan, with assistance from Du. The manuscript was written primarily by Parikh, along with Lairson, Morgan, and Du, and revised by Parikh.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/economics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biological Products/economics , Biological Products/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/economics , Aged , Aged, 80 and over , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/economics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/economics , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/economics , Bevacizumab/administration & dosage , Bevacizumab/economics , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/economics , Cost-Benefit Analysis , Female , Humans , Irinotecan , Male , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/economics , Oxaliplatin , Quality-Adjusted Life Years , Texas
12.
Value Health ; 19(4): 431-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27325335

ABSTRACT

BACKGROUND: Bevacizumab is used extensively in the treatment of cancer, including advanced ovarian cancer, for which results of the International Collaborative Ovarian Neoplasm (ICON) 7 trial have been recently reported. The National Institute for Health and Care Excellence's (NICE's) recent decision not to recommend bevacizumab for advanced ovarian cancer was not based on evidence related to the unlicensed lower dosage (7.5 mg/kg) of the drug despite its use in the English National Health Service (NHS) and the ICON7 trial. OBJECTIVE: To report on the findings of an analysis that considered whether the lower dose is cost-effective. METHODS: Cost-effectiveness analysis is assessed from the perspective of the English NHS and health outcomes expressed in terms of quality-adjusted life-years (QALYs). The analysis focuses on a clinically predefined high-risk subgroup of the ICON7 trial. The price at which the lower dose of bevacizumab could be considered cost-effective for the English NHS is presented for a range of scenarios to inform decisions about price negotiations by international health systems. RESULTS: In the base-case analysis, bevacizumab has an incremental cost-effectiveness ratio of £48,975 per additional QALY, which is above NICE's standard cost-effectiveness threshold (£20,000-£30,000 per QALY). The official price of bevacizumab in 2013 was between £2.31 and £2.63 per milligram. A price reduction of between 46% and 67%, dependent on the NICE threshold, would be required for the product to be cost-effective in the high-risk subgroup. CONCLUSIONS: The lower dose of bevacizumab for advanced ovarian cancer is not cost-effective based on the product's list price and using NICE's cost-effectiveness thresholds. Significant price discounts would be needed to make the drug affordable to the NHS.


Subject(s)
Angiogenesis Inhibitors/economics , Antineoplastic Combined Chemotherapy Protocols/economics , Bevacizumab/economics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/economics , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/economics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Phytogenic/economics , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/pharmacology , Carboplatin/economics , Carboplatin/therapeutic use , Cost-Benefit Analysis , Disease-Free Survival , Female , Humans , Paclitaxel/economics , Paclitaxel/therapeutic use , Quality-Adjusted Life Years , Severity of Illness Index , Survival Analysis
13.
Mem. Inst. Oswaldo Cruz ; 110(1): 1-22, 03/02/2015. tab, graf
Article in English | LILACS | ID: lil-741625

ABSTRACT

An increasingly asked question is 'can we confidently link bats with emerging viruses?'. No, or not yet, is the qualified answer based on the evidence available. Although more than 200 viruses - some of them deadly zoonotic viruses - have been isolated from or otherwise detected in bats, the supposed connections between bats, bat viruses and human diseases have been raised more on speculation than on evidence supporting their direct or indirect roles in the epidemiology of diseases (except for rabies). However, we are convinced that the evidence points in that direction and that at some point it will be proved that bats are competent hosts for at least a few zoonotic viruses. In this review, we cover aspects of bat biology, ecology and evolution that might be relevant in medical investigations and we provide a historical synthesis of some disease outbreaks causally linked to bats. We provide evolutionary-based hypotheses to tentatively explain the viral transmission route through mammalian intermediate hosts and to explain the geographic concentration of most outbreaks, but both are no more than speculations that still require formal assessment.


Subject(s)
Animals , Humans , Antineoplastic Agents, Phytogenic/isolation & purification , Antioxidants/isolation & purification , Fatty Acids/analysis , Industrial Waste/analysis , Malus/chemistry , Plant Oils/isolation & purification , Seeds/chemistry , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/economics , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/adverse effects , Antioxidants/economics , Antioxidants/pharmacology , Cell Line, Tumor , Chemical Phenomena , CHO Cells , Cricetulus , Cell Proliferation/drug effects , Dietary Supplements/adverse effects , Dietary Supplements/economics , Fatty Acids, Nonesterified/adverse effects , Fatty Acids, Nonesterified/analysis , Fatty Acids, Nonesterified/economics , Fatty Acids/adverse effects , Fatty Acids/economics , Food Preservatives/adverse effects , Food Preservatives/economics , Food Preservatives/isolation & purification , Food Preservatives/pharmacology , Food-Processing Industry/economics , Fruit/chemistry , Fruit/economics , India , Industrial Waste/economics , Linoleic Acid/adverse effects , Linoleic Acid/analysis , Linoleic Acid/economics , Oleic Acid/adverse effects , Oleic Acid/analysis , Oleic Acid/economics , Plant Oils/chemistry , Plant Oils/economics , Plant Oils/pharmacology
14.
Mol Nutr Food Res ; 58(10): 1970-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25045029

ABSTRACT

SCOPE: Maslinic acid is a bioactive minor component of Olea europaea L. with health-enhancing activities and no harmful effects. A pharmacokinetic (PK) study was conducted to determine its bioavailability for future studies of maslinic acid in humans. METHODS AND RESULTS: Intravenous (1 mg/kg) and oral (50 mg/kg) administrations to Sprague-Dawley rats were performed. Blood was obtained several times over 24 h and PKs were analyzed with NONMEM 7.2, applying a population approach. Body weight was included a priori in the model with fixed allometric exponents, based on allometric principles. Plasma concentrations versus time were best characterized by a two-open compartment model with first-order absorption and linear elimination. Maslinic acid had a relative rapid oral absorption with a peak concentration after administration at 0.51 h and a bioavailability of 5.13%. Once in bloodstream, it distributed extensively into tissues, since the central and peripheral distribution volumes were 8.41 L/70 kg and 63.6 L/70 kg, respectively. The clearance (8 L/h/70 kg) was related to unaltered renal excretion. The prediction-corrected visual predictive check confirmed its stability and predictive ability. CONCLUSION: An allometric population PK model was performed for maslinic acid, which adequately described and predicted plasma concentrations.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Fruit/chemistry , Models, Biological , Olea/chemistry , Triterpenes/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/blood , Antineoplastic Agents, Phytogenic/economics , Biological Availability , Body Weight , Dietary Supplements , Drug Stability , Food-Processing Industry/economics , Industrial Waste/analysis , Industrial Waste/economics , Injections, Intravenous , Intestinal Absorption , Male , Metabolic Clearance Rate , Rats, Sprague-Dawley , Renal Elimination , Tissue Distribution , Triterpenes/administration & dosage , Triterpenes/blood , Triterpenes/economics
15.
Eur J Cancer Prev ; 23(6): 568-78, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24911998

ABSTRACT

Several biological and physical factors confer complexity on cancer, which may be responsible for its associated morbidity. The successful management of this hyperproliferative disease is beyond the realm of therapeutics that may include the use of a single or at times a combination of treatment modalities. It is apparent that prevention with complex agents such as botanicals could serve as a possible potential solution. The present review focuses on the aspects of cancer that contribute toward making it an extremely complex disease and that may be tackled effectively with the use of 'multicomponent' botanicals.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Neoplasms/drug therapy , Neoplasms/etiology , Antineoplastic Agents, Phytogenic/economics , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Clone Cells/pathology , Complex Mixtures/therapeutic use , Drug Resistance, Neoplasm , Genetic Heterogeneity , Health Care Costs , Humans , Morbidity , Mortality , Neoplasms/economics , Neoplasms/epidemiology , Plant Extracts/therapeutic use
16.
J Sci Food Agric ; 94(7): 1373-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24114648

ABSTRACT

BACKGROUND: Large quantities of brewers' spent grain (BSG), a co-product of the brewing industry, are produced annually. BSG contains hydroxycinnamic acids, and phenolic-rich extracts from BSG have previously demonstrated the ability to protect against oxidant-induced DNA damage. The present study investigated the anti-inflammatory potential of eight phenolic extracts from BSG: four pale (P1-P4) and four black (B1-B4) extracts. RESULTS: BSG extracts were more cytotoxic in Jurkat T than U937 cells, with lower IC50 values in Jurkat T cells, measured using the (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Pale BSG extracts P2 and P3 showed the greatest anti-inflammatory potential, significantly (P < 0.05) reducing interleukin-2 (IL-2), interleukin-4 (IL-4, P2 only), interleukin-10 (IL-10) and interferon-γ (IFN-γ) production. In addition, extracts P1-P3 and B2-B4 showed significant (P < 0.05) antioxidant effects, determined by the cellular antioxidant activity assays superoxide dismutase, catalase and glutathione content (GSH). CONCLUSION: Phenolic extracts from BSG, particularly the pale BSG extracts, have the ability to reduce a stimulated cytokine production and may also protect against cellular oxidative stress. Results of the present study highlight the potential of BSG phenolic extracts to act as functional food ingredients, providing an alternative use and improving the value of this brewing industry co-product.


Subject(s)
Antioxidants/metabolism , Beer , Coumaric Acids/metabolism , Edible Grain/chemistry , Immunologic Factors/metabolism , Industrial Waste/analysis , Phenols/metabolism , Anti-Inflammatory Agents, Non-Steroidal/analysis , Anti-Inflammatory Agents, Non-Steroidal/economics , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antineoplastic Agents, Phytogenic/analysis , Antineoplastic Agents, Phytogenic/economics , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/metabolism , Antioxidants/analysis , Antioxidants/economics , Antioxidants/isolation & purification , Beer/economics , Beer/microbiology , Cell Line, Tumor , Coumaric Acids/analysis , Coumaric Acids/economics , Coumaric Acids/isolation & purification , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Edible Grain/economics , Food, Fortified/analysis , Food, Fortified/economics , Food-Processing Industry/economics , Humans , Immunologic Factors/analysis , Immunologic Factors/economics , Immunologic Factors/isolation & purification , Industrial Waste/economics , Ireland , Leukemia, T-Cell/immunology , Leukemia, T-Cell/metabolism , Monocytes/enzymology , Monocytes/immunology , Monocytes/metabolism , Phenols/analysis , Phenols/economics , Phenols/isolation & purification , Pigmentation , Plant Extracts/chemistry , Plant Extracts/economics , Plant Extracts/isolation & purification , Plant Extracts/metabolism , Recycling
17.
J Sci Food Agric ; 94(5): 929-34, 2014 Mar 30.
Article in English | MEDLINE | ID: mdl-23929365

ABSTRACT

BACKGROUND: Apple pomace is generated in huge quantities in juice-processing industries the world over and continuous efforts are being made for its inclusive utilization. In this study, apple seeds separated from industrial pomace were used for extraction of oil. The fatty acid composition, physicochemical and antioxidant as well as in vitro anticancer properties of extracted oil were studied to assess its suitability in food and therapeutic applications. RESULTS: The fatty acid composition of seed oil revealed the dominance of oleic (46.50%) and linoleic acid (43.81%). It had high iodine (121.8 g I 100 g⁻¹) and saponification value (184.91 mg KOH g⁻¹ oil). The acid value, refractive index and relative density were 4.28 mg KOH g⁻¹, 1.47 and 0.97 mg mL⁻¹, respectively. The antioxidant potential (IC50) of apple seed oil was 40.06 µg mL⁻¹. Cytotoxicity of apple seed oil against CHOK1, SiHa and A549 cancer cell lines ranged between 0.5 ± 0.06% and 88.6 ± 0.3%. CONCLUSION: The physicochemical properties of apple seed oil were comparable with edible food oil, indicating its better stability and broad application in the food and pharmaceutical industries. Apple seed oil could be a good source of natural antioxidants. Also, the in vitro cytotoxic activity against specific cell lines exhibited its potential as an anticancer agent.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Antioxidants/isolation & purification , Fatty Acids/analysis , Industrial Waste/analysis , Malus/chemistry , Plant Oils/isolation & purification , Seeds/chemistry , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/economics , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/adverse effects , Antioxidants/economics , Antioxidants/pharmacology , CHO Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Chemical Phenomena , Cricetulus , Dietary Supplements/adverse effects , Dietary Supplements/economics , Fatty Acids/adverse effects , Fatty Acids/economics , Fatty Acids, Nonesterified/adverse effects , Fatty Acids, Nonesterified/analysis , Fatty Acids, Nonesterified/economics , Food Preservatives/adverse effects , Food Preservatives/economics , Food Preservatives/isolation & purification , Food Preservatives/pharmacology , Food-Processing Industry/economics , Fruit/chemistry , Fruit/economics , Humans , India , Industrial Waste/economics , Linoleic Acid/adverse effects , Linoleic Acid/analysis , Linoleic Acid/economics , Oleic Acid/adverse effects , Oleic Acid/analysis , Oleic Acid/economics , Plant Oils/chemistry , Plant Oils/economics , Plant Oils/pharmacology
18.
Zhongguo Zhong Yao Za Zhi ; 39(20): 4050-3, 2014 Oct.
Article in Chinese | MEDLINE | ID: mdl-25751961

ABSTRACT

Risk monitoring of new Chinese patent anti-hepatoma drugs is tracking recognized risks and residual risks, identifying emerging risk and ensure the implementation of the plan, estimating the process of reducing effectiveness. The paper is mainly through understanding the status of Chinese patent anti-hepatoma drugs, the content, characteristic and analysis method of dynamic risk monitoring, and then select the risk control indicators, collect risk information. Finally, puts forward the thought of anti-hepatoma drugs listed evaluation in our country, and try to establish the model of dynamic risk management of anti-hepatoma drugs.


Subject(s)
Antineoplastic Agents, Phytogenic/economics , Carcinoma, Hepatocellular/drug therapy , Drug Discovery , Drugs, Chinese Herbal/economics , Liver Neoplasms/drug therapy , Product Surveillance, Postmarketing , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/therapeutic use , Drug Discovery/economics , Drug Discovery/legislation & jurisprudence , Drug Discovery/organization & administration , Drug and Narcotic Control/economics , Drug and Narcotic Control/legislation & jurisprudence , Drug and Narcotic Control/organization & administration , Drugs, Chinese Herbal/adverse effects , Drugs, Chinese Herbal/therapeutic use , Humans
19.
Expert Rev Pharmacoecon Outcomes Res ; 13(3): 381-91, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23534988

ABSTRACT

AIM: The COSTABRAX study evaluated the cost-effectiveness of nanoparticle albumin-bound paclitaxel (nab-paclitaxel) versus polyethylated castor oil-based standard paclitaxel (sb-paclitaxel) in the treatment of patients with previously treated metastatic breast cancer in Spain. MATERIALS & METHODS: Efficacy data were obtained from the CA012 trial (nab-paclitaxel administered every 3 weeks [q3w] and sb-paclitaxel q3w) and indirect comparison (sb-paclitaxel administered weekly), and were modeled to a time horizon of 5 years using a Markov model. The analysis was performed from the National Health Service perspective. Use of resources and key assumptions of the model were validated by a panel of 22 local oncologists. RESULTS: Compared with sb-paclitaxel q3w, nab-paclitaxel q3w was cost effective, with a cost per life year gained of €11,088 and a cost per quality-adjusted life year of €17,808. Compared with sb-paclitaxel administered weekly, it showed savings of €711 per patient. CONCLUSION: The COSTABRAX study showed that nab-paclitaxel q3w is a cost-effective alternative compared with sb-paclitaxel q3w and a cost-saving alternative to sb-paclitaxel administered weekly in Spain.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/drug therapy , Nanoparticles , Paclitaxel/therapeutic use , Albumin-Bound Paclitaxel , Albumins/administration & dosage , Albumins/economics , Albumins/therapeutic use , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/economics , Breast Neoplasms/economics , Breast Neoplasms/pathology , Castor Oil/analogs & derivatives , Castor Oil/chemistry , Cost Savings , Cost-Benefit Analysis , Drug Administration Schedule , Female , Humans , Markov Chains , Models, Economic , Neoplasm Metastasis , Paclitaxel/administration & dosage , Paclitaxel/economics , Quality-Adjusted Life Years , Spain , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...