%0 Book %A Institute of Medicine %E Weisfeld, Neil %E English, Rebecca A. %E Claiborne, Anne B. %T Envisioning a Transformed Clinical Trials Enterprise in the United States: Establishing an Agenda for 2020: Workshop Summary %@ 978-0-309-25315-4 %D 2012 %U https://nap.nationalacademies.org/catalog/13345/envisioning-a-transformed-clinical-trials-enterprise-in-the-united-states %> https://nap.nationalacademies.org/catalog/13345/envisioning-a-transformed-clinical-trials-enterprise-in-the-united-states %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 248 %X There is growing recognition that the United States' clinical trials enterprise (CTE) faces great challenges. There is a gap between what is desired - where medical care is provided solely based on high quality evidence - and the reality - where there is limited capacity to generate timely and practical evidence for drug development and to support medical treatment decisions. With the need for transforming the CTE in the U.S. becoming more pressing, the IOM Forum on Drug Discovery, Development, and Translation held a two-day workshop in November 2011, bringing together leaders in research and health care. The workshop focused on how to transform the CTE and discussed a vision to make the enterprise more efficient, effective, and fully integrated into the health care system. Key issue areas addressed at the workshop included: the development of a robust clinical trials workforce, the alignment of cultural and financial incentives for clinical trials, and the creation of a sustainable infrastructure to support a transformed CTE. This document summarizes the workshop. %0 Book %A Institute of Medicine %E Grossmann, Claudia %E Sanders, Julia %E English, Rebecca A. %T Large Simple Trials and Knowledge Generation in a Learning Health System: Workshop Summary %@ 978-0-309-28911-5 %D 2013 %U https://nap.nationalacademies.org/catalog/18400/large-simple-trials-and-knowledge-generation-in-a-learning-health-system %> https://nap.nationalacademies.org/catalog/18400/large-simple-trials-and-knowledge-generation-in-a-learning-health-system %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 118 %X Randomized clinical trials (RCTs) are often referred to as the "gold standard" of clinical research. However, in its current state, the U.S. clinical trials enterprise faces substantial challenges to the efficient and effective conduct of research. Streamlined approaches to RCTs, such as large simple trials (LSTs), may provide opportunities for progress on these challenges. Clinical trials support the development of new medical products and the evaluation of existing products by generating knowledge about safety and efficacy in pre- and post-marketing settings and serve to inform medical decision making and medical product development. Although well-designed and -implemented clinical trials can provide robust evidence, a gap exists between the evidence needs of a continuously learning health system, in which all medical decisions are based on the best available evidence, and the reality, in which the generation of timely and practical evidence faces significant barriers. Large Simple Trials and Knowledge Generation in a Learning Health System is the summary of a workshop convened by the Institute of Medicine's Roundtable on Value & Science-Driven Health Care and the Forum on Drug Discovery, Development, and Translation. Experts from a wide range of disciplines--including health information technology, research funding, clinical research methods, statistics, patients, product development, medical product regulation, and clinical outcomes research--met to marshal a better understanding of the issues, options, and approaches to accelerating the use of LSTs. This publication summarizes discussions on the potential of LSTs to improve the speed and practicality of knowledge generation for medical decision making and medical product development, including efficacy and effectiveness assessments, in a continuously learning health system. Large Simple Trials and Knowledge Generation in a Learning Health System explores acceleration of the use of LSTs to improve the speed and practicality of knowledge generation for medical decision making and medical product development; considers the concepts of LST design, examples of successful LSTs, the relative advantages of LSTs, and the infrastructure needed to build LST capacity as a routine function of care; identifies structural, cultural, and regulatory barriers hindering the development of an enhanced LST capacity; discusses needs and strategies in building public demand for and participation in LSTs; and considers near-term strategies for accelerating progress in the uptake of LSTs in the United States. %0 Book %A Institute of Medicine %T Developing a National Registry of Pharmacologic and Biologic Clinical Trials: Workshop Report %@ 978-0-309-10078-6 %D 2006 %U https://nap.nationalacademies.org/catalog/11561/developing-a-national-registry-of-pharmacologic-and-biologic-clinical-trials %> https://nap.nationalacademies.org/catalog/11561/developing-a-national-registry-of-pharmacologic-and-biologic-clinical-trials %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 124 %X To improve public confidence in clinical research, a number of public and private groups have called for a publicly accessible, comprehensive, and transparent registry of relevant information on clinical trials for drugs and biologics. The public and various entities within the medical community (health care providers, researchers, medical journal editors, pharmaceutical companies, health insurers, and regulators) have different expectations and perceived needs regarding a public clinical trial registry. The IOM Committee on Clinical Trial Registries hosted a workshop on June 27, 2005, to obtain much-needed input from members of the public, public advocate groups, and the broader community of journal editors, pharmaceutical and biotech leaders, NIH, and the FDA. Participants discussed the data elements that have been at the core of debate and commented on issues of compliance and implementation of a national clinical trial registry. Developing a National Registry of Pharmacologic and Biologic Clinical Trials: Workshop Report inlcudes discussions at the workshop centered on the following five concepts, and are described within this report: 1) Purpose, 2) Which Trials to Include, 3) Delayed Disclosure Mechanism, 4) Reporting Results of Completed Trials, and 5) Compliance. %0 Book %A Institute of Medicine %T Sharing Clinical Trial Data: Maximizing Benefits, Minimizing Risk %@ 978-0-309-31629-3 %D 2015 %U https://nap.nationalacademies.org/catalog/18998/sharing-clinical-trial-data-maximizing-benefits-minimizing-risk %> https://nap.nationalacademies.org/catalog/18998/sharing-clinical-trial-data-maximizing-benefits-minimizing-risk %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 304 %X Data sharing can accelerate new discoveries by avoiding duplicative trials, stimulating new ideas for research, and enabling the maximal scientific knowledge and benefits to be gained from the efforts of clinical trial participants and investigators. At the same time, sharing clinical trial data presents risks, burdens, and challenges. These include the need to protect the privacy and honor the consent of clinical trial participants; safeguard the legitimate economic interests of sponsors; and guard against invalid secondary analyses, which could undermine trust in clinical trials or otherwise harm public health. Sharing Clinical Trial Data presents activities and strategies for the responsible sharing of clinical trial data. With the goal of increasing scientific knowledge to lead to better therapies for patients, this book identifies guiding principles and makes recommendations to maximize the benefits and minimize risks. This report offers guidance on the types of clinical trial data available at different points in the process, the points in the process at which each type of data should be shared, methods for sharing data, what groups should have access to data, and future knowledge and infrastructure needs. Responsible sharing of clinical trial data will allow other investigators to replicate published findings and carry out additional analyses, strengthen the evidence base for regulatory and clinical decisions, and increase the scientific knowledge gained from investments by the funders of clinical trials. The recommendations of Sharing Clinical Trial Data will be useful both now and well into the future as improved sharing of data leads to a stronger evidence base for treatment. This book will be of interest to stakeholders across the spectrum of research—from funders, to researchers, to journals, to physicians, and ultimately, to patients. %0 Book %A Institute of Medicine %A National Academies of Sciences, Engineering, and Medicine %T Ovarian Cancers: Evolving Paradigms in Research and Care %@ 978-0-309-38046-1 %D 2016 %U https://nap.nationalacademies.org/catalog/21841/ovarian-cancers-evolving-paradigms-in-research-and-care %> https://nap.nationalacademies.org/catalog/21841/ovarian-cancers-evolving-paradigms-in-research-and-care %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %K Biology and Life Sciences %P 396 %X In an era of promising advances in cancer research, there are considerable and even alarming gaps in the fundamental knowledge and understanding of ovarian cancer. Researchers now know that ovarian cancer is not a single disease—several distinct subtypes exist with different origins, risk factors, genetic mutations, biological behaviors, and prognoses. However, persistent questions have impeded progress toward improving the prevention, early detection, treatment, and management of ovarian cancers. Failure to significantly improve morbidity and mortality during the past several decades is likely due to several factors, including the lack of research being performed by specific disease subtype, lack of definitive knowledge of the cell of origin and disease progression, and incomplete understanding of genetic and non-genetic risk factors. Ovarian Cancers examines the state of the science in ovarian cancer research, identifies key gaps in the evidence base and the challenges to addressing those gaps, considers opportunities for advancing ovarian cancer research, and examines avenues for translation and dissemination of new findings and communication of new information to patients and others. This study makes recommendations for public- and private-sector efforts that could facilitate progress in reducing the incidence of morbidity and mortality from ovarian cancers. %0 Book %A National Academies of Sciences, Engineering, and Medicine %E Addie, Siobhan %E Gee, Amanda Wagner %E Olson, Steve %E Beachy, Sarah H. %T Deriving Drug Discovery Value from Large-Scale Genetic Bioresources: Proceedings of a Workshop %@ 978-0-309-44778-2 %D 2016 %U https://nap.nationalacademies.org/catalog/23601/deriving-drug-discovery-value-from-large-scale-genetic-bioresources-proceedings %> https://nap.nationalacademies.org/catalog/23601/deriving-drug-discovery-value-from-large-scale-genetic-bioresources-proceedings %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 114 %X The process of discovering and developing a new drug or therapy is extremely costly and time consuming, and recently, it has been estimated that the creation of a new medicine costs on average more than $2 billion and takes 10 years to reach patients. The challenges associated with bringing new medicines to market have led many pharmaceutical companies to seek out innovative methods for streamlining their drug discovery research. One way to increase the odds of success for compounds in the drug development pipeline is to adopt genetically guided strategies for drug discovery, and recognizing the potential benefits of collecting genetic and phenotypic information across specific populations, pharmaceutical companies have started collaborating with healthcare systems and private companies that have curated genetic bioresources, or large databases of genomic information. Large-scale cohort studies offer an effective way to collect and store information that can be used to assess gene–environment interactions, identify new potential drug targets, understand the role of certain genetic variants in the drug response, and further elucidate the underlying mechanisms of disease onset and progression. To examine how genetic bioresources could be used to improve drug discovery and target validation, the National Academies of Sciences, Engineering, and Medicine hosted a workshop in March 2016. Participants at the workshop explored the current landscape of genomics-enabled drug discovery activities in industry, academia, and government; examined enabling partnerships and business models; and considered gaps and best practices for collecting population data for the purpose of improving the drug discovery process. This publication summarizes the presentations and discussions from the workshop. %0 Book %A Institute of Medicine %T Discussion Framework for Clinical Trial Data Sharing: Guiding Principles, Elements, and Activities %@ 978-0-309-29779-0 %D 2014 %U https://nap.nationalacademies.org/catalog/18610/discussion-framework-for-clinical-trial-data-sharing-guiding-principles-elements %> https://nap.nationalacademies.org/catalog/18610/discussion-framework-for-clinical-trial-data-sharing-guiding-principles-elements %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 72 %X Sharing data generated through the conduct of clinical trials offers the promise of placing evidence about the safety and efficacy of therapies and clinical interventions on a firmer basis and enhancing the benefits of clinical trials. Ultimately, such data sharing - if carried out appropriately - could lead to improved clinical care and greater public trust in clinical research and health care. Discussion Framework for Clinical Trial Data Sharing: Guiding Principles, Elements, and Activities is part of a study of how data from clinical trials might best be shared. This document is designed as a framework for discussion and public comment. This framework is being released to stimulate reactions and comments from stakeholders and the public. The framework summarizes the committee's initial thoughts on guiding principles that underpin responsible sharing of clinical trial data, defines key elements of clinical trial data and data sharing, and describes a selected set of clinical trial data sharing activities. %0 Book %A National Academies of Sciences, Engineering, and Medicine %E Addie, Siobhan %E Hackmann, Meredith %E Wizemann, Theresa %E Beachy, Sarah %T Implementing and Evaluating Genomic Screening Programs in Health Care Systems: Proceedings of a Workshop %@ 978-0-309-47341-5 %D 2018 %U https://nap.nationalacademies.org/catalog/25048/implementing-and-evaluating-genomic-screening-programs-in-health-care-systems %> https://nap.nationalacademies.org/catalog/25048/implementing-and-evaluating-genomic-screening-programs-in-health-care-systems %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 150 %X Genomic applications are being integrated into a broad range of clinical and research activities at health care systems across the United States. This trend can be attributed to a variety of factors, including the declining cost of genome sequencing and the potential for improving health outcomes and cutting the costs of care. The goals of these genomics-based programs may be to identify individuals with clinically actionable variants as a way of preventing disease, providing diagnoses for patients with rare diseases, and advancing research on genetic contributions to health and disease. Of particular interest are genomics- based screening programs, which will, in this publication, be clinical screening programs that examine genes or variants in unselected populations in order to identify individuals who are at an increased risk for a particular health concern (e.g., diseases, adverse drug outcomes) and who might benefit from clinical interventions. On November 1, 2017, the National Academies of Sciences, Engineering, and Medicine hosted a public workshop to explore the challenges and opportunities associated with integrating genomics-based screening programs into health care systems. This workshop was developed as a way to explore the challenges and opportunities associated with integrating genomics-based programs in health care systems in the areas of evidence collection, sustainability, data sharing, infrastructure, and equity of access. This publication summarizes the presentations and discussions from the workshop. %0 Book %A National Academies of Sciences, Engineering, and Medicine %E Wizemann, Theresa %E Gee, Amanda Wagner %E Shore, Carolyn %T Envisioning a Transformed Clinical Trials Enterprise for 2030: Proceedings of a Workshop %@ 978-0-309-26928-5 %D 2022 %U https://nap.nationalacademies.org/catalog/26349/envisioning-a-transformed-clinical-trials-enterprise-for-2030-proceedings-of %> https://nap.nationalacademies.org/catalog/26349/envisioning-a-transformed-clinical-trials-enterprise-for-2030-proceedings-of %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 138 %X The evolution of health care is expanding the possibilities for integration of clinical research into the continuum of clinical care; new approaches are enabling the collection of data in real-world settings; and new modalities, such as digital health technologies and artificial intelligence applications, are being leveraged to overcome challenges and advance clinical research. At the same time, the clinical research enterprise is strained by rising costs, varying global regulatory and economic landscapes, increasing complexity of clinical trials, barriers to recruitment and retention of research participants, and a clinical research workforce that is under tremendous demands. Looking ahead to 2030, the Forum on Drug Discovery, Development, and Translation of the National Academies of Sciences, Engineering, and Medicine convened a public workshop for stakeholders from across the drug research and development life cycle to reflect on the lessons learned over the past 10 years and consider opportunities for the future. The workshop was designed to consider goals and priority action items that could advance the vision of a 2030 clinical trials enterprise that is more efficient, effective, person-centered, inclusive, and integrated into the health care delivery system so that outcomes and experiences for all stakeholders are improved. This Proceedings of a Workshop summarizes the presentations and discussions that took place during the four-part virtual public workshop held on January 26, February 9, March 24, and May 11, 2021. %0 Book %A Institute of Medicine %E Nass, Sharyl J. %E Moses, Harold L. %E Mendelsohn, John %T A National Cancer Clinical Trials System for the 21st Century: Reinvigorating the NCI Cooperative Group Program %@ 978-0-309-15186-3 %D 2010 %U https://nap.nationalacademies.org/catalog/12879/a-national-cancer-clinical-trials-system-for-the-21st-century %> https://nap.nationalacademies.org/catalog/12879/a-national-cancer-clinical-trials-system-for-the-21st-century %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 316 %X The National Cancer Institute's (NCI) Clinical Trials Cooperative Group Program has played a key role in developing new and improved cancer therapies. However, the program is falling short of its potential, and the IOM recommends changes that aim to transform the Cooperative Group Program into a dynamic system that efficiently responds to emerging scientific knowledge; involves broad cooperation of stakeholders; and leverages evolving technologies to provide high-quality, practice-changing research. %0 Book %A Institute of Medicine %A National Academies of Sciences, Engineering, and Medicine %E Graig, Laurene A. %E Phillips, Jonathan K. %E Moses, Harold L. %T Biomarker Tests for Molecularly Targeted Therapies: Key to Unlocking Precision Medicine %@ 978-0-309-38134-5 %D 2016 %U https://nap.nationalacademies.org/catalog/21860/biomarker-tests-for-molecularly-targeted-therapies-key-to-unlocking-precision %> https://nap.nationalacademies.org/catalog/21860/biomarker-tests-for-molecularly-targeted-therapies-key-to-unlocking-precision %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 292 %X Every patient is unique, and the evolving field of precision medicine aims to ensure the delivery of the right treatment to the right patient at the right time. In an era of rapid advances in biomedicine and enhanced understanding of the genetic basis of disease, health care providers increasingly have access to advanced technologies that may identify molecular variations specific to an individual patient, which subsequently can be targeted for treatment. Known as biomarker tests for molecularly targeted therapies, these complex tests have the potential to enable the selection of the most beneficial treatment (and also to identify treatments that may be harmful or ineffective) for the molecular underpinnings of an individual patient's disease. Such tests are key to unlocking the promise of precision medicine. Biomarker tests for molecularly targeted therapies represent a crucial area of focus for developing methods that could later be applicable to other areas of precision medicine. The appropriate regulatory oversight of these tests is required to ensure that they are accurate, reliable, properly validated, and appropriately implemented in clinical practice. Moreover, common evidentiary standards for assessing the beneficial impact of biomarker-guided therapy selection on patient outcomes, as well as the effective collection and sharing of information related to those outcomes, are urgently needed to better inform clinical decision making. Biomarker Tests of Molecularly Targeted Therapies examines opportunities for and challenges to the use of biomarker tests to select optimal therapy and offers recommendations to accelerate progress in this field. This report explores regulatory issues, reimbursement issues, and clinical practice issues related to the clinical development and use of biomarker tests for targeting therapies to patients. Properly validated, appropriately implemented biomarker tests hold the potential to enhance patient care and improve outcomes, and therefore addressing the challenges facing such tests is critical. %0 Book %A Institute of Medicine %E Mack, Alison %E Nass, Sharyl J. %T Implementing a National Cancer Clinical Trials System for the 21st Century: Workshop Summary %@ 978-0-309-21268-7 %D 2011 %U https://nap.nationalacademies.org/catalog/13154/implementing-a-national-cancer-clinical-trials-system-for-the-21st-century %> https://nap.nationalacademies.org/catalog/13154/implementing-a-national-cancer-clinical-trials-system-for-the-21st-century %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 122 %X Clinical trials enable scientific discoveries to advance patient care, in addition to informing and guiding subsequent research. The National Cancer Institute's (NCI's) Clinical Trials Cooperative Group Program works to advance patient care and research. The Cooperative Group Program has been instrumental in establishing the standards for cancer patient care and clinical research methods. Despite broad participation in the program, financial strain and procedural burdens limit the ability of the Cooperative Group Program to undertake medical practice-changing clinical research. Thus, the Institute of Medicine's (IOM's) National Cancer Policy Forum and the American Society of Clinical Oncology held a workshop on March 21, 2011 to follow up on the 2010 IOM report, A National Clinical Trials System for the 21st Century: Reinvigorating the NCI Cooperative Group Program, which made recommendations to strengthen the NCI Cooperative Group Program. In keeping with the established commitment to excellence Implementing a National Cancer Clinical Trials System for the 21st Century outlines how to improve the current system by incorporating innovative science and trial design into cancer clinical trials. It also examines the impact of increasing quality in regards to speed, efficiency, design, launch, and conduct, as well as improving prioritization, and incentivized participation. %0 Book %A National Academies of Sciences, Engineering, and Medicine %E Gee, Amanda Wagner %E Balogh, Erin %E Patlak, Margie %E Nass, Sharyl J. %T The Drug Development Paradigm in Oncology: Proceedings of a Workshop %@ 978-0-309-45794-1 %D 2018 %U https://nap.nationalacademies.org/catalog/24742/the-drug-development-paradigm-in-oncology-proceedings-of-a-workshop %> https://nap.nationalacademies.org/catalog/24742/the-drug-development-paradigm-in-oncology-proceedings-of-a-workshop %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 144 %X Advances in cancer research have led to an improved understanding of the molecular mechanisms underpinning the development of cancer and how the immune system responds to cancer. This influx of research has led to an increasing number and variety of therapies in the drug development pipeline, including targeted therapies and associated biomarker tests that can select which patients are most likely to respond, and immunotherapies that harness the body’s immune system to destroy cancer cells. Compared with standard chemotherapies, these new cancer therapies may demonstrate evidence of benefit and clearer distinctions between efficacy and toxicity at an earlier stage of development. However, there is a concern that the traditional processes for cancer drug development, evaluation, and regulatory approval could impede or delay the use of these promising cancer treatments in clinical practice. This has led to a number of efforts—by patient advocates, the pharmaceutical industry, and the Food and Drug Administration (FDA)—to accelerate the review of promising new cancer therapies, especially for cancers that currently lack effective treatments. However, generating the necessary data to confirm safety and efficacy during expedited drug development programs can present a unique set of challenges and opportunities. To explore this new landscape in cancer drug development, the National Academies of Sciences, Engineering, and Medicine developed a workshop held in December 2016. This workshop convened cancer researchers, patient advocates, and representatives from industry, academia, and government to discuss challenges with traditional approaches to drug development, opportunities to improve the efficiency of drug development, and strategies to enhance the information available about a cancer therapy throughout its life cycle in order to improve its use in clinical practice. This publication summarizes the presentations and discussions from the workshop. %0 Book %A National Research Council %T The Prevention and Treatment of Missing Data in Clinical Trials %@ 978-0-309-15814-5 %D 2010 %U https://nap.nationalacademies.org/catalog/12955/the-prevention-and-treatment-of-missing-data-in-clinical-trials %> https://nap.nationalacademies.org/catalog/12955/the-prevention-and-treatment-of-missing-data-in-clinical-trials %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %K Behavioral and Social Sciences %K Surveys and Statistics %P 162 %X Randomized clinical trials are the primary tool for evaluating new medical interventions. Randomization provides for a fair comparison between treatment and control groups, balancing out, on average, distributions of known and unknown factors among the participants. Unfortunately, these studies often lack a substantial percentage of data. This missing data reduces the benefit provided by the randomization and introduces potential biases in the comparison of the treatment groups. Missing data can arise for a variety of reasons, including the inability or unwillingness of participants to meet appointments for evaluation. And in some studies, some or all of data collection ceases when participants discontinue study treatment. Existing guidelines for the design and conduct of clinical trials, and the analysis of the resulting data, provide only limited advice on how to handle missing data. Thus, approaches to the analysis of data with an appreciable amount of missing values tend to be ad hoc and variable. The Prevention and Treatment of Missing Data in Clinical Trials concludes that a more principled approach to design and analysis in the presence of missing data is both needed and possible. Such an approach needs to focus on two critical elements: (1) careful design and conduct to limit the amount and impact of missing data and (2) analysis that makes full use of information on all randomized participants and is based on careful attention to the assumptions about the nature of the missing data underlying estimates of treatment effects. In addition to the highest priority recommendations, the book offers more detailed recommendations on the conduct of clinical trials and techniques for analysis of trial data. %0 Book %A Institute of Medicine %E Nass, Sharyl J. %E Patlak, Margie %T Implementing a National Cancer Clinical Trials System for the 21st Century: Second Workshop Summary %@ 978-0-309-28724-1 %D 2013 %U https://nap.nationalacademies.org/catalog/18362/implementing-a-national-cancer-clinical-trials-system-for-the-21st-century %> https://nap.nationalacademies.org/catalog/18362/implementing-a-national-cancer-clinical-trials-system-for-the-21st-century %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 110 %X The National Clinical Trials Network (NCTN) supported by the National Cancer Institute (NCI) has played an integral role in cancer research and in establishing the standard of care for cancer patients for more than 50 years. Formerly known as the NCI Clinical Trials Cooperative Group Program, the NCTN is comprised of more than 2,100 institutions and 14,000 investigators, who enroll more than 20,000 cancer patients in clinical trials each year across the United States and internationally. Recognizing the recent transformative advances in cancer research that necessitate modernization in how cancer clinical trials are run, as well as inefficiencies and other challenges impeding the national cancer clinical trials program, the NCI asked the IOM to develop a set of recommendations to improve the federally funded cancer clinical trials system. These recommendations were published in the 2010 report, A National Cancer Clinical Trials System for the 21st Century: Reinvigorating the NCI Cooperative Group Program. In early 2011, the NCPF and the American Society of Clinical Oncology (ASCO) held a workshop in which stakeholders discussed the changes they planned to implement in response to the IOM goals and recommendations. Two years later, on February 11-12, 2013, in Washington, DC, the NCPF and ASCO reconvened stakeholders to report on the changes they have made thus far to address the IOM recommendations. At this workshop, representatives from the NCI, the NCTN, comprehensive cancer centers, patient advocacy groups, the Food and Drug Administration (FDA), industry, and other stakeholders highlighted the progress that has been made in achieving the goals for a reinvigorated national cancer clinical trials system. Implementing a National Cancer Clinical Trials System for the 21st Century is a summary of that workshop. %0 Book %A Institute of Medicine %E Pray, Leslie %E Robinson, Sally %T Challenges for the FDA: The Future of Drug Safety: Workshop Summary %@ 978-0-309-10986-4 %D 2007 %U https://nap.nationalacademies.org/catalog/11969/challenges-for-the-fda-the-future-of-drug-safety-workshop %> https://nap.nationalacademies.org/catalog/11969/challenges-for-the-fda-the-future-of-drug-safety-workshop %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 128 %X As the principal agency regulating food, drugs, medical devices, and biological products used by Americans, the U.S. Food and Drug Administration (FDA) serves one of the most critical consumer protection functions of the federal government. The FDA's reach is enormous, regulating products that represent roughly 25 percent of all consumer spending in the United States. Since 1992, however, federal funding for the agency has diminished, and the FDA's Center for Drug Evaluation and Research (CDER) currently relies on the fees it receives from the industry it regulates to fund the majority of its drug regulation functions. Prescription drug safety is receiving heightened press coverage and congressional scrutiny as a result of recent, highly publicized events, such as the recall of Vioxx because of its link to heart attacks, and the link between certain antidepressants (selective serotonin reuptake inhibitors, or SSRIs) and an increased risk of suicidal ideation in children. To address these concerns, the FDA in 2005 commissioned the Institute of Medicine (IOM) to conduct an independent assessment of the current U.S. drug safety system. In September 2006, the IOM committee released its report—The Future of Drug Safety: Promoting and Protecting the Health of the Public—which included 25 recommendations for improving the system for drug safety review. The committee identified four major vulnerabilities in the U.S. drug safety system: (1) chronic underfunding; (2) organization problems, particularly inadequate integration of pre-and postmarket data review; (3) a range of technical problems related to the insufficient quantity and quality of postmarket data and inadequate capability to systematically monitor the risks and benefits of drugs after marketing; and (4) unclear regulatory authority and insufficiently flexible regulatory tools. Since the IOM report was issued, the FDA has taken a number of steps toward implementing the recommended improvements. Like many government agencies, however, the FDA is financially strained by its existing responsibilities, and fully implementing the recommended improvements to the drug safety system would require significant financial commitments.The IOM report addressed some of the costs associated with its recommendations, but left many unanswered questions about the resources required to fully achieve the envisioned improvements. To better understand the types and magnitude of resources required to achieve the goals of the IOM report, the IOM's Forum on Drug Discovery, Development, and Translation convened a 1-day symposium in March 2007. Challenges for the FDA: The Future of Drug Safety, Workshop Summary explains the presentations and discussions in seven key areas: addressing the FDA's resource challenges; strengthening the scientific base of the agency; integrating pre- and postmarket review; enhancing postmarket safety monitoring; conducting confirmatory drug safety and efficacy studies; enhancing the value of clinical trial registration; and enhancing the FDA's postmarket regulation and enforcement. %0 Book %A Institute of Medicine %E Wizemann, Theresa %E Robinson, Sally %E Giffin, Robert %T Breakthrough Business Models: Drug Development for Rare and Neglected Diseases and Individualized Therapies: Workshop Summary %@ 978-0-309-12088-3 %D 2009 %U https://nap.nationalacademies.org/catalog/12219/breakthrough-business-models-drug-development-for-rare-and-neglected-diseases %> https://nap.nationalacademies.org/catalog/12219/breakthrough-business-models-drug-development-for-rare-and-neglected-diseases %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %P 150 %X The process for developing new drug and biologic products is extraordinarily expensive and time-consuming. Although large pharmaceutical companies may be able to afford the cost of development because they can expect a large return on investment, organizations developing drugs to treat rare and neglected diseases are unable to rely on such returns. On June 23, 2008, the Institute of Medicine's Forum on Drug Discovery, Development, and Translation held a public workshop, "Breakthrough Business Models: Drug Development for Rare and Neglected Diseases and Individualized Therapies," which sought to explore new and innovative strategies for developing drugs for rare and neglected diseases. %0 Book %A Institute of Medicine %E Olson, Steve %E Downey, Autumn S. %T Sharing Clinical Research Data: Workshop Summary %@ 978-0-309-26874-5 %D 2013 %U https://nap.nationalacademies.org/catalog/18267/sharing-clinical-research-data-workshop-summary %> https://nap.nationalacademies.org/catalog/18267/sharing-clinical-research-data-workshop-summary %I The National Academies Press %C Washington, DC %G English %K Health and Medicine %K Policy for Science and Technology %P 156 %X Pharmaceutical companies, academic researchers, and government agencies such as the Food and Drug Administration and the National Institutes of Health all possess large quantities of clinical research data. If these data were shared more widely within and across sectors, the resulting research advances derived from data pooling and analysis could improve public health, enhance patient safety, and spur drug development. Data sharing can also increase public trust in clinical trials and conclusions derived from them by lending transparency to the clinical research process. Much of this information, however, is never shared. Retention of clinical research data by investigators and within organizations may represent lost opportunities in biomedical research. Despite the potential benefits that could be accrued from pooling and analysis of shared data, barriers to data sharing faced by researchers in industry include concerns about data mining, erroneous secondary analyses of data, and unwarranted litigation, as well as a desire to protect confidential commercial information. Academic partners face significant cultural barriers to sharing data and participating in longer term collaborative efforts that stem from a desire to protect intellectual autonomy and a career advancement system built on priority of publication and citation requirements. Some barriers, like the need to protect patient privacy, pre- sent challenges for both sectors. Looking ahead, there are also a number of technical challenges to be faced in analyzing potentially large and heterogeneous datasets. This public workshop focused on strategies to facilitate sharing of clinical research data in order to advance scientific knowledge and public health. While the workshop focused on sharing of data from preplanned interventional studies of human subjects, models and projects involving sharing of other clinical data types were considered to the extent that they provided lessons learned and best practices. The workshop objectives were to examine the benefits of sharing of clinical research data from all sectors and among these sectors, including, for example: benefits to the research and development enterprise and benefits to the analysis of safety and efficacy. Sharing Clinical Research Data: Workshop Summary identifies barriers and challenges to sharing clinical research data, explores strategies to address these barriers and challenges, including identifying priority actions and "low-hanging fruit" opportunities, and discusses strategies for using these potentially large datasets to facilitate scientific and public health advances.