Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Lancet Oncol ; 25(4): 431-438, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38547890

RESUMEN

BACKGROUND: The financial impact of cancer medicines on health systems is not well known. We describe temporal trends in expenditure on cancer medicines within the single-payer health system of Ontario, Canada, and the extent of clinical benefit these treatments offer. METHODS: In this cross-sectional study, we identified cancer medicines and expenditures from formularies and costing databases (the New Drug Funding Program, Ontario Drug Benefit Program, and The High-Cost Therapy Funding Program) during 10 consecutive years (April 1, 2012, to March 31, 2022) in Ontario, Canada. For intravenous medicines, we applied the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS) to identify expenditures associated with substantial clinical benefit. We also identified treatments associated with improved overall survival or quality of life. FINDINGS: 69 intravenous and 98 oral or injectable medicines were funded during 2012-22. Annual expenditure on cancer medicines increased by approximately 15% per year during 2012-22; the increase was more rapid in the most recent 4 years. Total expenditure on cancer medicines in the 2021-22 financial year was CA$1·7 billion. Immune checkpoint inhibitors were the single biggest expense by class ($284 million), representing 17% of the entire cancer medicine annual budget. Drugs with the highest individual costs were lenalidomide ($178 million) and pembrolizumab ($163 million), each accounting for around 10% of the entire budget. 29 (76%) of 38 indications eligible for ESMO-MCBS scoring met the threshold for substantial clinical benefit. Eight (21%) indications had no randomised trial evidence of improved overall survival, and only four (11%) were associated with improved QOL. $346 million (67% of the expenditure on intravenous cancer medicines) was spent on drugs that improved median overall survival by more than 6 months, $82 million (16%) was spent on medicines with overall survival gains of 3-6 months, and $32 million (6%) was spent on medicines with overall survival gains of less than 3 months. $53 million (10%) was spent on medicines with no established improvement in overall survival. INTERPRETATION: Costs of cancer medicines to the Canadian health system are increasing rapidly. Most funded indications met thresholds for substantial clinical benefit and two-thirds of the expenditure were for medicines that improve survival by more than 6 months. Whether this cost trajectory can be maintained in a sustainable, equitable, high-quality health system is unclear. Efforts are needed to ensure the price of medicines with substantial benefit is affordable and funding of treatments with very modest benefit might need to be re-assessed, particularly when alternative supportive and palliative therapies are available. FUNDING: None.


Asunto(s)
Neoplasias , Calidad de Vida , Humanos , Estudios Transversales , Ontario , Salud Pública , Neoplasias/tratamiento farmacológico
2.
Lancet Oncol ; 20(8): 1065-1069, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31296490

RESUMEN

BACKGROUND: The fragility index of trial results-ie, the minimum number of changes from non-events to events resulting in loss of statistical significance-can provide a measure of confidence that a positive effect reported in a randomised controlled trial is real. We aimed to calculate the fragility index of randomised controlled trials supporting US Food and Drug Administration (FDA)-approved anticancer drugs. METHODS: This is a retrospective analysis of phase 3, randomised, controlled trials supporting anticancer drugs that were approved by the FDA between Jan 1, 2014, and Dec 31, 2018. Two-arm studies with 1:1 randomisation and significant positive results for a time-to-event outcome were eligible for the fragility index calculation, which involves the iterative addition of an event to the experimental group (defined as the group with the smaller number of events in positive trials) and concomitant subtraction of a non-event from that group, until positive significance (defined as p<0·05 by Fisher's exact test) is lost. FINDINGS: We identified 36 phase 3 randomised controlled trials, of which 17 (47%) were included in the fragility index analysis. The median fragility index was 2 (IQR 0-27). The fragility index was 2 or less in nine (53%) of 17 trials; for these trials, the fragility index was 1% or less of the total sample size. In five (29%) of 17 trials, the number lost to follow-up was more than the fragility index. INTERPRETATION: Many phase 3 randomised controlled trials supporting FDA-approved anticancer drugs have a low fragility index, challenging confidence for concluding their superiority over control treatments. Although not a measure of effect, the fragility index might provide an additional means of assessing the robustness of clinical trial data. FUNDING: None.


Asunto(s)
Antineoplásicos/uso terapéutico , Ensayos Clínicos Fase III como Asunto , Neoplasias/tratamiento farmacológico , Ensayos Clínicos Controlados Aleatorios como Asunto , Ensayos Clínicos Fase III como Asunto/métodos , Ensayos Clínicos Fase III como Asunto/normas , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Ensayos Clínicos Controlados Aleatorios como Asunto/normas , Estudios Retrospectivos
3.
Lancet Oncol ; 18(7): 887-894, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28583794

RESUMEN

BACKGROUND: The American Society of Clinical Oncology (ASCO) and the European Society for Medical Oncology (ESMO) have developed frameworks that quantify survival gains in light of toxicity and quality of life to assess the benefits of cancer therapies. We applied these frameworks to a cohort of contemporary randomised controlled trials to explore agreement between the two approaches and to assess the relation between treatment benefit and cost. METHODS: We identified all randomised controlled trials of systemic therapies in non-small-cell lung cancer, breast cancer, colorectal cancer, and pancreatic cancer published between Jan 1, 2011, and Dec 31, 2015, and assessed their abstracts and methods. Trials were eligible for inclusion in our cohort if significant differences favouring the experimental group in a prespecified primary or secondary outcome were reported (secondary outcomes were assessed only if primary outcomes were not significant). We assessed trial endpoints with the ASCO and ESMO frameworks at two timepoints 3 months apart to confirm intra-rater reliability. Cohen's κ statistic was calculated to establish agreement between the two frameworks on the basis of the median ASCO score, which was used as an arbitrary threshold of benefit, and the framework-recommended ESMO threshold. Differences in monthly drug cost between the experimental and control groups of each randomised controlled trial (ie, incremental drug cost) were derived from 2016 average wholesale prices. FINDINGS: 109 randomised controlled trials were eligible for inclusion, 42 (39%) in non-small-cell lung cancer, 36 (33%) in breast cancer, 25 (23%) in colorectal cancer, and six (6%) in pancreatic cancer. ASCO scores ranged from 2 to 77; median score was 25 (IQR 16-35). 41 (38%) trials met the benefit thresholds in the ESMO framework. Agreement between the two frameworks was fair (κ=0·326). Among the 100 randomised controlled trials for which drug costing data were available, ASCO benefit score and monthly incremental drug costs were negatively correlated (ρ=-0·207; p=0·039). Treatments that met ESMO benefit thresholds had a lower median incremental drug cost than did those that did not meet benefit thresholds (US$2981 [IQR 320-9059] vs $8621 [1174-13 930]; p=0·018). INTERPRETATION: There is only fair correlation between these two major value care frameworks, and negative correlations between framework outputs and drug costs. Delivery of optimal cancer care in a sustainable health system will necessitate future oncologists, investigators, and policy makers to reconcile the disconnect between drug cost and clinical benefit. FUNDING: None.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Colorrectales/tratamiento farmacológico , Análisis Costo-Beneficio/métodos , Costos de los Medicamentos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Antineoplásicos/economía , Antineoplásicos/uso terapéutico , Humanos , Calidad de Vida , Ensayos Clínicos Controlados Aleatorios como Asunto , Reproducibilidad de los Resultados , Estudios Retrospectivos , Tasa de Supervivencia
4.
Intern Med J ; 47(5): 586-588, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28503874

RESUMEN

First described in 1959, intravascular lymphoma (IVL) remains one of the most clinically challenging diagnoses due to its diverse and non-specific clinical manifestations and evasiveness in detection by standard investigations. Indeed, IVL deserves the title of 'medicine's greatest imitator'. We highlight a case of IVL where the diagnosis came too late in the clinical course, detected by random skin biopsy. Clinicians should strongly consider this diagnosis in presentations with persistent symptomatology despite appropriate interventions.


Asunto(s)
Flujo Sanguíneo Regional/fisiología , Grasa Subcutánea/irrigación sanguínea , Grasa Subcutánea/patología , Neoplasias Vasculares/diagnóstico , Neoplasias Vasculares/fisiopatología , Anciano , Velocidad del Flujo Sanguíneo/fisiología , Diagnóstico Diferencial , Femenino , Humanos , Linfoma de Células B Grandes Difuso/diagnóstico , Linfoma de Células B Grandes Difuso/fisiopatología
5.
Pflugers Arch ; 468(1): 43-58, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26395471

RESUMEN

The mammalian homeostatic oxygen sensing system (HOSS) initiates changes in vascular tone, respiration, and neurosecretion that optimize oxygen uptake and tissue oxygen delivery within seconds of detecting altered environmental or arterial PO2. The HOSS includes carotid body type 1 cells, adrenomedullary cells, neuroepithelial bodies, and smooth muscle cells (SMCs) in pulmonary arteries (PAs), ductus arteriosus (DA), and fetoplacental arteries. Hypoxic pulmonary vasoconstriction (HPV) optimizes ventilation-perfusion matching. In utero, HPV diverts placentally oxygenated blood from the non-ventilated lung through the DA. At birth, increased alveolar and arterial oxygen tension dilates the pulmonary vasculature and constricts the DA, respectively, thereby transitioning the newborn to an air-breathing organism. Though modulated by endothelial-derived relaxing and constricting factors, O2 sensing is intrinsic to PASMCs and DASMCs. Within the SMC's dynamic mitochondrial network, changes in PO2 alter the reduction-oxidation state of redox couples (NAD(+)/NADH, NADP(+)/NADPH) and the production of reactive oxygen species, ROS (e.g., H2O2), by complexes I and III of the electron transport chain (ETC). ROS and redox couples regulate ion channels, transporters, and enzymes, changing intracellular calcium [Ca(2+)]i and calcium sensitivity and eliciting homeostatic responses to hypoxia. In PASMCs, hypoxia inhibits ROS production and reduces redox couples, thereby inhibiting O2-sensitive voltage-gated potassium (Kv) channels, depolarizing the plasma membrane, activating voltage-gated calcium channels (CaL), increasing [Ca(2+)]i, and causing vasoconstriction. In DASMCs, elevated PO2 causes mitochondrial fission, increasing ETC complex I activity and ROS production. The DASMC's downstream response to elevated PO2 (Kv channel inhibition, CaL activation, increased [Ca(2+)]i, and rho kinase activation) is similar to the PASMC's hypoxic response. Impaired O2 sensing contributes to human diseases, including pulmonary arterial hypertension and patent DA.


Asunto(s)
Conducto Arterial/metabolismo , Pulmón/metabolismo , Mitocondrias Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Canales de Calcio/metabolismo , Humanos , Pulmón/irrigación sanguínea , Oxígeno/sangre , Canales de Potasio/metabolismo
6.
8.
Sci Rep ; 13(1): 16138, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37752147

RESUMEN

Patients endure risk and uncertainty when they participate in clinical trials. We previously estimated that 12,217 patient-participants are required to bring a new cancer drug to market. However, many development efforts are aimed at extending the label of already approved drugs. Herein, we estimate the number of patients required to extend the indication of an FDA approved cancer drug. We identified all anti-cancer drugs approved by the FDA 2012 to 2015. We searched clinicaltrials.gov to identify all drug development trajectories (i.e., a series of one or more clinical trials testing a unique drug-indication pairing) launched after FDA approval for each drug. We identified which trajectories produced the following milestones: secondary FDA approvals, secondary FDA approvals achieving substantial clinical benefit in ESMO-MCBS, and recommendations in NCCN clinical practice guidelines. Using the total enrollment, we estimated the number of patients needed to reach each milestone. Forty-two drugs were approved by the FDA between 2012 and 2015, leading to 451 post-approval trajectories enrolling 129,548 patients. Fourteen secondary FDA approvals were identified, of which 4 met the ESMO-MCBS definition of substantial clinical benefit. Fourteen NCCN off-label recommendations were obtained. A total of 9253, 32,387 and 4627 patients were needed to attain an FDA approval, an approval with substantial clinical benefit on ESMO-MCBS, and an NCCN guideline recommendation, respectively. The number of patients needed to obtain a first secondary FDA approval was 16,596. Large numbers of patients are needed to extend the label of prior FDA approved drugs. Label extension after approval entails lower marginal costs for developers. However, extra knowledge available to researchers about a drug's safety and pharmacology after FDA approval does not appear to translate into reduced patient numbers required for developing new cancer applications.


Asunto(s)
Neoplasias , Humanos , Estudios Retrospectivos , Neoplasias/tratamiento farmacológico , Desarrollo de Medicamentos , Conocimiento , Pacientes
9.
J Clin Epidemiol ; 158: 134-140, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37028684

RESUMEN

OBJECTIVES: Drug developers sometimes launch phase 3 (P3) trials without supporting evidence from phase 2 (P2) trials. We call this practice "P2 bypass." The aims of this study were to estimate the prevalence of P2 bypass and to compare the safety and efficacy results for P3 trials that bypassed with those that did not. STUDY DESIGN AND SETTING: We created a sample of P3 solid tumor trials registered on ClinicalTrials.gov with primary completion dates between 2013 and 2019. We then attempted to match each with a supporting P2 trial using strict and broad criteria. P3 outcomes were meta-analyzed using a random effects model with subgroup contrast between trials that bypassed and those that did not. RESULTS: 129 P3 trial arms met eligibility and nearly half involved P2 bypass. P3 trials involving P2 bypass produced significantly and nonsignificantly worse pooled efficacy estimates using broad and strict matching criteria, respectively. We did not observe significant differences in safety outcomes between P3 trials that bypassed P2 and those that did not. CONCLUSION: The risk/benefit balance of P3 trials that bypassed P2 is less favourable than for trials supported by P2.


Asunto(s)
Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Ensayos Clínicos Fase III como Asunto , Ensayos Clínicos Fase II como Asunto
10.
Curr Oncol ; 29(4): 2530-2538, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35448181

RESUMEN

Background: In this study, we compared and contrasted design characteristics, results, and publications of randomized controlled trials (RCTs) in gastrointestinal (GI), lung, and breast cancer. Methods: A PUBMED search identified phase III RCTs of anticancer therapy in GI, lung, and breast cancer published globally during the period 2014−2017. Descriptive statistics, chi-square tests, and the Kruskal−Wallis test were used to compare RCT design, results, and output across the cancer sites. Results: A total of 352 RCTs were conducted on GI (36%), lung (29%), and breast (35%) cancer. Surrogate endpoints were used in 55% of trials; this was most common in breast trials (72%) compared to GI (47%) and lung trials (43%, p < 0.001). Breast trials more often met their primary endpoint (54%) than GI (41%) and lung trials (41%) (p = 0.024). When graded with the ESMO-MCBS, lung cancer trials (50%, 15/30) were more likely to meet the threshold for substantial benefit. GI trials were published in journals with a substantially lower impact factor (IF; median IF 13) than lung (median IF 21) and breast cancer trials (median IF 21) (p = 0.038). Conclusions: Important differences in RCT design and output exist between the three major cancer sites. Use of surrogate endpoints and the magnitude of benefit associated with new treatments vary substantially across cancer sites.


Asunto(s)
Neoplasias de la Mama , Neoplasias Pulmonares , Neoplasias de la Mama/terapia , Femenino , Humanos , Pulmón , Neoplasias Pulmonares/tratamiento farmacológico , Oncología Médica , Ensayos Clínicos Controlados Aleatorios como Asunto , Investigación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA