28 research outputs found

    Improving value of clinical research - an evidence-based approach

    Get PDF
    Evidence suggests that 85% of biomedical research spending, i.e. 200 billion US dollars every year, goes to waste. In 2014, The Lancet published a series of five reviews showing how dividends from the investment in research might be increased at five stages - from the relevance and priorities of the questions being asked, to how the research is designed, conducted, and reported. Value and waste have since then become buzzwords in the academic as well as public debate surrounding health research. Although academic institutions are the major driving force of patient-oriented clinical research receiving large proportions of public funding they have been slow responders to the Series’ recommendations. Some of the identified underlying reasons include a lack of a common understanding of “value” as a concept and sparse practical guidance for academia on how to improve it. This work represents the first effort to formulate an academic response to The Lancet series on increasing value in clinical research by investigating the two distinct concepts in the equation: “Quality” and “cost”. In a first step, we systematically reviewed existing quality concepts, both in the medical literature and across international clinical research stakeholder groups. Precise definitions of quality were sparse, and stakeholder perspectives of crucial components of quality varied. Based on these findings, we then engaged international stakeholder representatives in the creation of a comprehensive, consensus-based framework for the quality of clinical research that is applicable to all study types and spans the entire lifecycle of a clinical study, i.e. from conceptualization of the research question to dissemination of study results. Primarily, it is designed to be operationalized in the academic setting and fully supports the REWARD Statement. This framework builds the foundation for a common understanding of the concept of “quality” and its practical assessment. At Swiss national level, the framework has triggered all stakeholders to convene in a first symposium on how to increase value of academic clinical research and serves as an agenda for future research on research. In a second step, we systematically reviewed the current evidence on the costs and associated resource use of Randomized Controlled Trials (RCTs), which we found to be sparse. Based on this, we laid the foundation for future study cost assessments in academia by (i) developing a comprehensive list of items for the retrospective and prospective assessment of costs, and (ii) generating first empirical evidence on main cost drivers in a case report on two academic RCTs. Although these two RCTs were conducted in very different settings and resulted in vastly different costs, the main drivers, i.e. personnel costs during conduct phase, were the same. In addition, we investigated the added value of two innovative aspects that affect both study quality and cost, i.e. risk-based trial monitoring and remote data collection. We show that both concepts may increase the cost-effectiveness of trial conduct and thereby increase value, but only if the methodology is further investigated and then, rigorously implemented. Although we did not take the initially envisioned cost-consequence approach, we have certainly created awareness on value and waste in the academic context and engaged the major stakeholders in fundamental discussions on how to improve the current situation. In the future, the costs occurred need to inform quality assessments of clinical studies in order to create a tool that creates “value”, rather than sole quality conformity. Furthermore, the willingness-to-pay of academic decision makers in resource-constrained settings will weigh into the value equation and needs further investigation in the future. The impact of this work - and whether it eventually increases value in the system - now critically depends on its rigorous implementation, evaluation, and refinement

    How to conceptualize and implement a PhD program in health sciences - the Basel approach

    Get PDF
    Over the past decade, several excellent guidelines have been published on how to enhance the quality of PhD education in Europe. Aimed primarily at preparing students for innovative roles in their fields, they include variously structured approaches to curricular offerings, as well as other program components applicable across specialties (eg: supervisor support, scientific conduct, transferable skills). Since 2012, the interdisciplinary PhD Program in Health Sciences (PPHS) at the Faculty of Medicine of the University of Basel in Switzerland has focused on translating these guidelines into a 21st-century health sciences PhD program.; The PPHS started in 2012 based on the European Union (EU) guidelines for PhD education. This article describes the resulting interdisciplinary PhD program's conceptual underpinnings, rationale, structures, and 10 building blocks, like student portfolios, thematic training, interdisciplinary research seminars, student-initiated interdisciplinary activities, financial support of course participation, top-up and extension stipends, PhD supervision, research integrity, alumni follow-up network, and promotional tools including a dedicated website. Students enter from Clinical Research, Medicine Development, Nursing Science, Epidemiology and Public Health including Insurance Medicine, Sport Science (all from the Faculty of Medicine), and Epidemiology (Faculty of Science).; The Basel PPHS exemplifies state-of-the-art PhD education in Health Sciences based on European guidelines and offers guidance to other groups from conceptualization to rollout of an interdisciplinary health sciences PhD program

    Validity of mobile electronic data capture in clinical studies: a pilot study in a pediatric population.

    Get PDF
    BACKGROUND: Clinical studies in children are necessary yet conducting multiple visits at study centers remains challenging. The success of "care-at-home" initiatives and remote clinical trials suggests their potential to facilitate conduct of pediatric studies. This pilot aimed to study the feasibility of remotely collecting valid (i.e. complete and correct) saliva samples and clinical data utilizing mobile technology. METHODS: Single-center, prospective pilot study in children undergoing elective tonsillectomy at the University of Basel Children's Hospital. Data on pain scores and concomitant medication and saliva samples were collected by caregivers on two to four inpatient study days and on three consecutive study days at home. A tailored mobile application developed for this study supported data collection. The primary endpoint was the proportion of complete and correct caregiver-collected data (pain scale) and saliva samples in the at-home setting. Secondary endpoints included the proportion of complete and correct saliva samples in the inpatient setting, subjective feasibility for caregivers, and study cost. RESULTS: A total number of 23 children were included in the study of which 17 children, median age 6.0 years (IQR 5.0, 7.4), completed the study. During the at-home phase, 71.9% [CI = 64.4, 78.6] of all caregiver-collected pain assessments and 53.9% [CI = 44.2, 63.4] of all saliva samples were complete and correct. Overall, 64.7% [CI = 58.7, 70.4] of all data collected by caregivers at home was complete and correct. The predominant reason for incorrectness of data was adherence to the timing of predefined patient actions. Participating caregivers reported high levels of satisfaction and willingness to participate in similar trials in the future. Study costs for a potential sample size of 100 patients were calculated to be 20% lower for the at-home than for a traditional in-patient study setting. CONCLUSIONS: Mobile device supported studies conducted at home may provide a cost-effective approach to facilitate conduct of clinical studies in children. Given findings in this pilot study, data collection at home may focus on electronic data capture rather than biological sampling

    Reporting quality of clinical trial protocols: a repeated cross-sectional study about the Adherence to SPIrit Recommendations in Switzerland, CAnada and GErmany (ASPIRE-SCAGE)

    Full text link
    OBJECTIVES Comprehensive protocols are key for the planning and conduct of randomised clinical trials (RCTs). Evidence of low reporting quality of RCT protocols led to the publication of the Standard Protocol Items: Recommendations for Interventional Trials (SPIRIT) checklist in 2013. We aimed to examine the quality of reporting of RCT protocols from three countries before and after the publication of the SPIRIT checklist. DESIGN Repeated cross sectional study. SETTING Swiss, German and Canadian research ethics committees (RECs). PARTICIPANTS RCT protocols approved by RECs in 2012 (n=257) and 2016 (n=292). PRIMARY AND SECONDARY OUTCOME MEASURES The primary outcomes were the proportion of reported SPIRIT items per protocol and the proportion of trial protocols reporting individual SPIRIT items. We compared these outcomes in protocols approved in 2012 and 2016, and built regression models to explore factors associated with adherence to SPIRIT. For each protocol, we also extracted information on general trial characteristics and assessed whether individual SPIRIT items were reported RESULTS: The median proportion of reported SPIRIT items among RCT protocols showed a non-significant increase from 72% (IQR, 63%-79%) in 2012 to 77% (IQR, 68%-82%) in 2016. However, in a preplanned subgroup analysis, we detected a significant improvement in investigator-sponsored protocols: the median proportion increased from 64% (IQR, 55%-72%) in 2012 to 76% (IQR, 64%-83%) in 2016, while for industry-sponsored protocols median adherence was 77% (IQR 72%-80%) for both years. The following trial characteristics were independently associated with lower adherence to SPIRIT: single-centre trial, no support from a clinical trials unit or contract research organisation, and investigator-sponsorship. CONCLUSIONS In 2012, industry-sponsored RCT protocols were reported more comprehensively than investigator-sponsored protocols. After publication of the SPIRIT checklist, investigator-sponsored protocols improved to the level of industry-sponsored protocols, which did not improve

    De novo fatty acid synthesis by Schwann cells is essential for peripheral nervous system myelination

    Get PDF
    Myelination calls for a remarkable surge in cell metabolism to facilitate lipid and membrane production. Endogenous fatty acid (FA) synthesis represents a potentially critical process in myelinating glia. Using genetically modified mice, we show that Schwann cell (SC) intrinsic activity of the enzyme essential for de novo FA synthesis, fatty acid synthase (FASN), is crucial for precise lipid composition of peripheral nerves and fundamental for the correct onset of myelination and proper myelin growth. Upon FASN depletion in SCs, epineurial adipocytes undergo lipolysis, suggestive of a compensatory role. Mechanistically, we found that a lack of FASN in SCs leads to an impairment of the peroxisome proliferator-activated receptor (PPAR) γ–regulated transcriptional program. In agreement, defects in myelination of FASN-deficient SCs could be ameliorated by treatment with the PPARγ agonist rosiglitazone ex vivo and in vivo. Our results reveal that FASN-driven de novo FA synthesis in SCs is mandatory for myelination and identify lipogenic activation of the PPARγ transcriptional network as a putative downstream functional mediator

    Reporting quality of trial protocols improved for non-regulated interventions but not regulated interventions: A repeated cross-sectional study

    Get PDF
    Objectives: To investigate the adherence of randomised controlled trial (RCT) protocols evaluating non-regulated interventions (including dietary interventions, surgical procedures, behavioural and lifestyle interventions, and exercise programmes) in comparison with regulated interventions to the Standard Protocol Items: Recommendations for Interventional Trials (SPIRIT) 2013 Statement. Methods: We conducted a repeated cross-sectional investigation in a random sample of RCT protocols approved in 2012 (n = 257) or 2016 (n = 292) by research ethics committees in Switzerland, Germany, or Canada. We investigated the proportion of accurately reported SPIRIT checklist items in protocols of trials with non-regulated as compared to regulated interventions. Results: Overall, 131 (24%) of trial protocols tested non-regulated interventions. In 2012, the median proportion of SPIRIT items reported in these protocols (59%, interquartile range [IQR], 53%-69%) was lower than in protocols with regulated interventions (median, 74%, IQR, 66%-80%). In 2016, the reporting quality of protocols with non-regulated interventions (median, 75%, IQR, 62%-83%) improved to the level of regulated intervention protocols, which had not changed on average. Conclusions: Reporting of RCT protocols evaluating non-regulated interventions improved between 2012 and 2016, although remained suboptimal. SPIRIT recommendations need to be further endorsed by researchers, ethics committees, funding agencies, and journals to optimize reporting of RCT protocols

    Reporting quality of trial protocols improved for non-regulated interventions but not regulated interventions: A repeated cross-sectional study

    No full text
    OBJECTIVES To investigate the adherence of randomised controlled trial (RCT) protocols evaluating non-regulated interventions (including dietary interventions, surgical procedures, behavioural and lifestyle interventions, and exercise programmes) in comparison with regulated interventions to the Standard Protocol Items: Recommendations for Interventional Trials (SPIRIT) 2013 Statement. METHODS We conducted a repeated cross-sectional investigation in a random sample of RCT protocols approved in 2012 (n = 257) or 2016 (n = 292) by research ethics committees in Switzerland, Germany, or Canada. We investigated the proportion of accurately reported SPIRIT checklist items in protocols of trials with non-regulated as compared to regulated interventions. RESULTS Overall, 131 (24%) of trial protocols tested non-regulated interventions. In 2012, the median proportion of SPIRIT items reported in these protocols (59%, interquartile range [IQR], 53%-69%) was lower than in protocols with regulated interventions (median, 74%, IQR, 66%-80%). In 2016, the reporting quality of protocols with non-regulated interventions (median, 75%, IQR, 62%-83%) improved to the level of regulated intervention protocols, which had not changed on average. CONCLUSIONS Reporting of RCT protocols evaluating non-regulated interventions improved between 2012 and 2016, although remained suboptimal. SPIRIT recommendations need to be further endorsed by researchers, ethics committees, funding agencies, and journals to optimize reporting of RCT protocols

    Academic response to improving value and reducing waste: A comprehensive framework for INcreasing QUality In patient-oriented academic clinical REsearch (INQUIRE)

    No full text
    <div><p>Background</p><p>Compelling evidence has demonstrated that a large proportion of investment in biomedical research is wasted; this waste is avoidable. Academic institutions have, thus far, shown limited response to recommendations for increasing value and reducing waste. We formulated an academic response by (i) achieving consensus across a wide range of stakeholder groups on a comprehensive framework for quality of patient-oriented clinical research and (ii) highlighting first successful examples of its operationalization to facilitate waste-reducing strategies at academic institutions.</p><p>Methods and findings</p><p>Based on a systematic review of quality definitions, concepts, and criteria in the medical literature (systematic MEDLINE search up to February 15, 2015, with independent and in duplicate article selection) and on stakeholder websites from 13 countries (Australia, Austria, Canada, France, Germany, Italy, Japan, Norway, Spain, Sweden, Switzerland, United Kingdom, and United States), we systematically developed a comprehensive framework for the quality of clinical research. We identified websites through personal contacts with experts in clinical research or public health who also suggested, for each country, websites of the following 7 stakeholder groups: patient organizations; academic research infrastructures; governmental bodies; regulatory agencies; ethics committees; the pharmaceutical industry; and funding agencies. In addition, we searched websites of inter- or supranational bodies involved in clinical research until no further insights emerged. After consolidation of the identified definitions, concepts, and criteria of quality in a basic framework structure, we conducted 4 rounds of an adapted online Delphi process among the same 7 stakeholder groups from 16 countries. The Delphi process ultimately achieved consensus on structure and content. The framework addresses 5 study stages (concept, planning and feasibility, conduct, analysis and interpretation, and reporting and knowledge translation) and includes the following dimensions: (i) protection of patient safety and rights, (ii) relevance/patient centeredness and involvement, (iii) minimization of bias (internal validity), (iv) precision, (v) transparency/access to data, and (vi) generalizability (external validity) of study results. These dimensions interact with 2 promoters—infrastructure and sustainability through education—that include a set of factors that may enhance all listed quality dimensions. Each quality dimension contains specific questions and explanatory items that guide quality assessment at each research stage from conceptualization of the research question through reporting and knowledge translation of study results. In the last survey round, Delphi participants from 9 countries (Austria, Australia, Canada, Germany, Italy, the Netherlands, Switzerland, UK, and US) agreed on the structure, content, and wording of the research stages, quality dimensions, specific questions, and descriptive examples of the final framework. In Switzerland, INQUIRE has resulted in a roadmap that guides initiatives to increase value within the Swiss Clinical Trial Organization network and through affiliated researchers.</p><p>Conclusions</p><p>We present a framework based on a consensus of different stakeholder groups guiding the practical assessment of clinical research quality at all stages of a research project. Operationalization of this common structure will support the increase of value by guiding academic institutions and researchers in developing quality enhancement initiatives, from posing the right research question to the transparent publication of results.</p></div

    SCTO roadmap on how to increase value in Swiss academic clinical research.

    No full text
    <p><sup>1</sup>The CTU Network and SCTO actively support the design, planning, and conduct of investigator-initiated studies that apply for the Swiss National Science Foundation’s call for investigator-initiated clinical trials (<a href="http://www.snf.ch/en/funding/programmes/iict/Pages/default.aspx" target="_blank">http://www.snf.ch/en/funding/programmes/iict/Pages/default.aspx</a>). The Swiss National Science Foundation has earmarked CHF 10 million for the 2017 funding period. Performance measures would allow assessing the SCTO network’s impact on the success of supported studies. <sup>2</sup>SCTO platforms are excellence clusters for specific support units, e.g., data management, statistics, training, and education, located at different CTUs. CTU, Clinical Trial Unit; REWARD, Reduce Research Waste and Reward Diligence.</p

    INQUIRE’s specific quality questions (and descriptive examples for Stage I) by quality dimension and research stage, and promoters.

    No full text
    <p>INQUIRE’s specific quality questions (and descriptive examples for Stage I) by quality dimension and research stage, and promoters.</p
    corecore