36
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Progress in Rare Diseases Research 2010–2016: An IRDiRC Perspective

      review-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Rare diseases by definition are conditions that affect small numbers of people. The prevalence threshold that designates a disease as rare varies in different countries. Generally, diseases with prevalence fewer than 5 in 10,000 people are considered rare.1 Many rare diseases affect far fewer people worldwide, with some having a single identified case and others with cases numbering from tens to low hundreds of people.2 However, collectively, they are common. There are ∼6,000–8,000 rare diseases, with 250–280 new diseases described annually, affecting an estimated 6–8% of the human population.3, 4, 5, 6 The rarity of these diseases individually creates significant challenges for affected patients, their families, and for clinicians attempting to achieve a confirmed diagnosis and implement best care.7, 8 Obtaining a correct diagnosis is frequently a difficult and lengthy process, as physicians and caregivers often lack appropriate expertise in a disease that they rarely encounter.4 Effective therapeutics are lacking for the large majority of these diseases, and when they exist, they are often very expensive because of scientific and manufacturing challenges, and small potential markets for such products; this creates barriers to access that are frequently difficult to resolve.3 Rare diseases are chronic and often severely disabling, thus treating these patients places a substantial burden on healthcare budgets. For example, one recent study from Western Australia concluded that in 2010 the state population affected by a limited cohort of only 467 rare diseases represented 2% of the population but 10.5% of in‐patient hospital costs.9 Therefore, improved diagnostics and targeted therapeutics that keep these patients healthier and reduce their time in medical facilities would be highly beneficial. Many rare diseases resemble common ones and involve the same genetic pathways, but are generally more aggressive or severe in their presentation. Improved understanding of these diseases may therefore be relevant to improving or developing diagnostics and therapeutics for their more common counterparts. Most rare diseases are genetic in their etiology, thus research addressing them systematically begins with efforts to identify genetic variants that are causative for particular diseases.1, 5 Such research has been under way for decades and has been successful, with many links between genetic mutations and diseases identified. However, by the mid‐2000s it became increasingly apparent that several factors were limiting success and needed to be addressed systematically on an international scale. Linking a genetic variant with a particular disease requires finding two or, preferably, more unrelated individuals who have a genetic variant in common, as well as exhibiting similar phenotypic characteristics. Identifying such matches can be facilitated by data sharing, but due to at times competitive academic framework and limitations in the consent obtained from research participants, data sharing among research groups working on the same disease was often limited or nonexistent. An additional problem was the lack of a standardized vocabulary to describe phenotypes, further complicating the identification of patients with similar symptoms.10 At the same time, the advent of next‐generation genomic techniques, which dramatically increased throughput and lowered cost, provided the opportunity to drastically accelerate the discovery of gene–disease connections.11 However, with this promise came the additional technical challenges of storing, analyzing, sending, and comparing large sets of data. The introduction of regulatory and economic incentives (e.g., market exclusivity, fee waivers, protocol assistance) was also successful in attracting the biopharmaceutical industry, which was previously reluctant to invest in rare disease therapeutic development with little or limited return on investment.5 Prior to the introduction of the Orphan Drug Act in the US in 1983, only 10 orphan drugs were approved in a decade, compared with 247 drugs for over 200 rare diseases in the course of the 25 years postlegislation.12 In Europe, following the adoption of the European Commission (EC) Regulation 141/2000, 63 orphan medicinal products received marketing authorization by 2010, a decade after implementation.13 While a large increase in the number of orphan designations was seen on both sides of the Atlantic, the average combined number of orphan‐designated products approved was about 15 per year and they addressed only a handful of rare diseases. By 2010, ∼400 orphan medicinal products were available for less than 300 rare diseases. Additionally, observations by the US Food and Drug Administration (FDA) of a slowdown instead of the expected acceleration in innovative medical therapies reaching patients were also reported in the mid‐2000s.14 Attrition rate aside, therapy development for rare diseases faces a number of specific challenges, including small populations for clinical studies, difficulty in determining relevant outcome measures and endpoints, and poorly understood natural histories. The heterogeneous pathophysiology and dispersed nature of rare diseases means research and development efforts and patient populations are scattered around the world. A scarcity of expertise poses a huge challenge to patients who seek access to diagnostic testing and appropriate treatment. In this overall setting, following initial discussions between Dr Ruxandra Draghia‐Akli of the Directorate‐General for Research and Innovation (DG RTD) of the European Commission (EC) and Dr Francis Collins of the US National Institutes of Health (NIH), the International Rare Diseases Research Consortium (IRDiRC) was formed.15 A consortium that unites public and private sector funders of research, patient advocacy groups, and scientific researchers, IRDiRC is committed to a global partnership to advance rare diseases research. This is delivered by attracting further resources to the domain and facilitating better international collaboration and coordination among research groups. In 2011, at its inauguration, IRDiRC set two ambitious goals for the rare diseases research collective—to develop 200 new therapies and the means to diagnose most rare genetic diseases by the year 2020. Six years after its official launch, IRDiRC is reflecting on its progress and achievements toward these goals, and considers how best to move forward even more ambitiously in the coming years. In this context, we review the history and the key success factors, as they provide the best markers for our future success and overcoming the apparently insurmountable issues ahead for people living with rare diseases, rare diseases stakeholders, industry, and policy makers. HISTORY The EC's DG RTD and the US NIH met in 2009 to discuss the need for expanded and further integrated efforts to address the global imperative for governments to collect public health data and provide solutions for patients affected with rare diseases. A preparatory workshop held in Reykjavík, Iceland, in October 2010 led to the creation of IRDiRC.15 Formally launched in 2011 to foster international research collaboration and investment in the field, IRDiRC had two aspirational goals: i) to contribute to the development of 200 new therapies, and ii) to develop the means to diagnose most rare diseases by the year 2020. At its launch, five public research funders committed to the IRDiRC mission. By the end of 2011, IRDiRC had expanded to 18 public funding members in addition to three international umbrella patient groups. The first private funders, i.e., companies, joined the effort in 2012. The Consortium has since expanded globally to include nearly 50 members across Europe, North America, Asia, Australia, and the Middle East (Figure 1), each—excepting the umbrella patient groups—committed to invest a minimum of USD 10 million in rare diseases research funding over 5 years.16 Through this global reach, with the cooperation of its public and private funders and patient groups, IRDiRC began to develop common scientific and policy frameworks to guide research and development in rare diseases under the leadership of Dr Ruxandra Draghia‐Akli (Chair, 2011–2012), Dr Paul Lasko (Chair, 2013–2015), and Dr Christopher Austin (Chair, 2016–present). The work of IRDiRC and its committees is critical for the development of new rare diseases knowledge, which is in turn vital for governments to develop informed, collaborative, and evidence‐based policy, and for academics and industry to be guided in the development of new diagnostics and therapies for rare diseases. Figure 1 IRDiRC member institutions are found across Europe, North America, Middle East, Asia, and Australia. Its membership has grown steadily over the years, with participation from research funders, companies, and patient advocates. While IRDiRC has a wide range of members, it is recognized that expertise in rare diseases, just as patients, is distributed around the world. In order to engage with international stakeholders in rare diseases research inside and outside of the Consortium, to assess the progress on the IRDiRC objectives, and to share acquired knowledge and competences, three international IRDiRC Conferences have been organized, to date. These conferences, held in Dublin, Ireland; Shenzhen, China; and Paris, France, respectively, advanced discussions on rare diseases research, and emphasized the utmost importance of collaborations among the global rare disease community to progress and success. GOVERNANCE OF IRDiRC IRDiRC is currently governed through a Consortium Assembly (formerly Executive Committee), three Scientific Committees, three Constituent Committees, an Operating Committee, ad hoc Task Forces, and a Scientific Secretariat (Figure 2). The Consortium Assembly is composed of one representative per IRDiRC member, and the Chairs and Vice Chairs of each of the three Scientific Committees. The Consortium Assembly acts as the primary forum to exchange information on issues that influence IRDiRC goals and activities, coordinates scientific and policy efforts in addressing research priorities to advance IRDiRC goals, adopts policies and guidelines, and provides strategic vision to foster rare diseases research. IRDiRC members of the Consortium Assembly further subdivide into three Constituent Committees, one each for Funders, Companies, and Patient Advocates, which each identify common priorities, roadblocks, and gaps across their respective constituent space, and coordinate activities to address those needs. The three Scientific Committees, one each for Diagnostics, Interdisciplinary, and Therapies, advise the Consortium Assembly on research priorities and progress made from a scientific viewpoint, identify bottlenecks and funding gaps in rare diseases research, and agree on actions in their scientific areas that will bring IRDiRC closer to its goals. The Operating Committee—consisting of the Chairs and Vice Chairs of the Consortium Assembly, the Constituent Committees and the Scientific Committees, and the Scientific Secretariat—is the key working committee that monitors and manages progress of IRDiRC activities and processes, provides information to the Consortium Assembly, and enables a more effective management of the Consortium. In order to push policy changes forward and provide technical solutions to specific, prioritized topics, ad hoc Task Forces—each consisting of topically relevant experts—are proposed and set up by the Scientific and Constituent Committees. The organizational, secretarial, reporting, and communications support to IRDiRC and its various members are provided by the Scientific Secretariat. Together, the Consortium contributes to the acceleration of rare diseases research, diagnostics and therapeutics development, reinforces international research cooperation and coordination, and maximizes the impact of investments made in the field of rare diseases research, all in order to bring diagnoses and therapies to rare disease patients throughout the world. Figure 2 IRDiRC is composed of a Consortium Assembly (formerly Executive Committee), an Operating Committee, three Constituent Committees, three Scientific Committees, a number of Task Forces, and a Scientific Secretariat. OBJECTIVES AND PROGRESS ON OBJECTIVES 2011–2020, NOW 2017 To meet IRDiRC's goals, collaborative actions and overarching policies are needed to address the challenges encountered as well as streamlining access to relevant data and patient populations. Funding Research in rare diseases is urgently needed, as many rare disease patients still lack an accurate and confirmed diagnosis and appropriate treatment. Funding of rare diseases research should not be carried out in isolation; appropriate coordinated effort through a network of funders will ensure that the impact of research investment is maximized through deduplication of research projects, generation of knowledge in lesser studied diseases, creation of common frameworks and infrastructures to facilitate data and patient sample sharing, and translation of basic research into preclinical and clinical studies. Collaborative funding mechanisms also enable the financing of large, transnational projects that require resources beyond what can be provided by individual, national research programs. The IRDiRC Scientific Secretariat, in collaboration with Orphanet,17 collects information pertaining to the projects funded by IRDiRC members. Data from IRDiRC public research funding members (excluding data from the NIH National Cancer Institute, from which data collection is ongoing) illustrate the financing of over 3,000 projects since 2010. A majority of them are basic and preclinical research projects, accounting for about 87% of funded projects, while clinical research (including observational studies and clinical trials) represents ∼8% of the total number of awarded projects, and epidemiological, health economics and social sciences studies account for 4% of funded research (Figure 3). Collectively, the basic and preclinical research projects covered nearly 1,200 rare diseases, while the clinical trials covered ∼220 rare diseases. The former mainly focused on rare neurological diseases (including neuromuscular diseases) and developmental disorders (including conditions associated with dysmorphology and congenital anomalies), whereas clinical trials were mainly oriented towards rare cancers and rare neurological diseases (Figures 4, 5). The prevalence and intersection of these disorder groupings in diagnostic and therapeutic domains reflect the biological and clinical overlaps between developmental disorders and cancer.18 This also reflects the reciprocal opportunities between and within rare syndromic diseases, rare cancers, and their intersections for insights into new therapies and for drug repurposing.19 Drug clinical trials constitute the overwhelming majority of trials (Figure  6), with more than half of them in earlier phases of development (phases I to II), 31% in phase III and 5% in phase IV (Figure 7). Approximately half of all clinical trials were funded by companies (48%), the others supported by public institutions. These data serve as the foundation for much of the new work of the Constituent Committees aimed at identifying areas of need, prioritizing, and addressing those gaps via coordinated implementation activities,20 while taking into consideration the promising21 bedside‐to‐bench translational approach to increase the likelihood of success in drug development and clinical translation. Figure 3 Distribution of funded projects and trials by type of research. Figure 4 Distribution of research projects by medical domain. Figure 5 Distribution of clinical trials by medical domain. Figure 6 Distribution of clinical trials by modality. Figure 7 Distribution of drug clinical trials by phase. Data received from public funders also demonstrated strong research investments and increasing number of collaborative calls on rare diseases, a sign of global commitment toward advancing rare diseases research and thus a commitment to bring diagnoses and treatments to rare disease patients. The EC, an IRDiRC cofounder, through its Framework Programme 7 (2007–2013) and Horizon 2020 Programme (2014–2020), invested around €762M in 135 rare diseases projects between 2010 and 2015,22 and expect to finance more projects in the coming years. The US NIH, another IRDiRC cofounder, has invested around US$10B between 2011 and 2016 via seven institutes and centers that are member organizations of IRDiRC, and the National Center for Advancing Translational Sciences’ Office of Rare Diseases Research (ORDR) alone has invested over US$100M in around 180 rare diseases projects between 2010 and 2015. E‐Rare, a joint transnational funding network, grew from 15 funding partners in 2010 to 26 funding partners in 2017.23 Between 2011 and 2016, E‐Rare funded 77 projects with a budget of €67.5M;24 the largest research funding call took place in 2015 when 19 projects were financed through an investment of €19.2M. Funders at the national level play an equally important role in financing rare diseases research. As an example, in Spain, the National Institute of Health Carlos III spent a total of €43.4M in the period of 2010–2016 to finance around 45–60 projects each year, while additionally earmarking an average of €5M per year to support the Centre of Biomedical Research Network for Rare Diseases (CIBERER). The Japan Agency for Medical Research and Development (AMED), established in April 2015, set up a dedicated Division of Rare/Intractable Disease Research. In 2 years, AMED has disbursed ¥20B (€153M/US$176M) and funded over 350 research projects. The Canadian Institutes of Health Research's (CIHR) Institute of Genetics has invested over CA$110M between 2011 and 2016. In recent years, patient organizations have also been making impressive investments in rare diseases research in order to develop diagnostic tools and therapeutics for their patients. Telethon Italia, for example, funded 337 extramural research projects in 2010–2016, as well as other intramural research activities and support initiatives, for a total budget close to €235M. A comprehensive data collection and analysis of rare diseases research funding, including the number and types of projects, and number and categories of rare diseases studied, for 2010–2016, is currently under way; the report is expected to be published in late 2017 to complement the State of Art of Research in Rare Diseases report. Progress in Diagnostics and Therapeutics Advances in diagnostics and therapeutics in the rare disease space over the past 6 years are attributed to a combination of factors and many players. IRDiRC monitors a number of metrics to quantify the progress made towards its key objectives. Progress in the diagnostics field relies on the identification of rare diseases and their underlying cause, often genetic in origin, and the development and availability of associated analytical tests. In this setting, the IRDiRC Diagnostics and Interdisciplinary Scientific Committees, in collaboration with Orphanet, analyzed the number of diseases for which a clinical test is available; at present, a genetic test is available for around 3,600 rare diseases, compared with 2,200 in 2010. Based on data curated by Orphanet and Online Mendelian Inheritance in Man (OMIM), an average of about 260–280 rare genetic diseases are discovered per year.4 In recent years, the majority of novel genes were discovered through exome sequencing approaches, with decreased use of previous discovery methods such as positional cloning. However, the discovery rate of exome sequencing over the past couple of years appears to be declining—this may be attributed to the fact that most of the simpler, monogenic rare diseases that are tractable to current techniques have been discovered, leaving the more complex cases to be solved. Nonetheless, in the last 5 years diagnostic efficiency has increased from ∼10% to 30–50%, a considerable step forward for rare disease patients.25 Yet, a number of bottlenecks remain to be tackled to continue the pace of discoveries and classification of disease‐related genes and variants. In the past two decades, several countries have introduced policies dedicated to orphan drug development and others are currently following suit with specific policies for their jurisdictions.26 To better grasp the number of therapies available for rare disease patients, IRDiRC tracks the cumulative number of approvals of medicinal products with a marketing authorization and orphan designation since 2010 via data from the European Medicines Agency (EMA) and the FDA. An increase has been observed in the number of drug approvals that reach the market in both Europe and the United States, and more are expected to come, as the number of orphan designations has steadily risen, as well. Between 2010 and 2016, over 220 new drugs have reached the market, covering over 170 diseases. The observed rise in the number of orphan drug designations holds a tremendous promise for the future of rare disease drug development, and ultimately for rare disease patients. With the addition of new members, and in particular Japan in 2015, IRDiRC aims to capture data from specific geographic‐regulatory regions. Thus, from 2017 onward, IRDiRC will be tracking approvals by the Pharmaceuticals and Medical Devices Agency (PMDA) in Japan and more globally as other significant regulatory jurisdictions are added to the Consortium. IRDiRC Policies and Guidelines In order to i) improve collaborations in rare diseases research, ii) increase the involvement of patients and their representatives in all relevant aspects of research, and iii) improve the sharing of data and resources, IRDiRC has developed a set of principles that address such common roadblocks. The “IRDiRC Policies and Guidelines” were developed by the Scientific Committees to serve as recommendations for research funders and researchers to provide guidance on key topics that include ontologies, diagnostics, biomarkers, patient registries, biobanks, natural history studies, therapeutics, models, publications, intellectual property, and communication.27 These policies and guidelines have been adopted by the IRDiRC members themselves through their own intramural rare diseases research and extramural rare diseases research funding programs, and provide guidelines for the entire rare diseases research community.27, 28 Addressing Barriers and Bottlenecks Barriers and bottlenecks in diagnostic and therapeutic research prolong the odyssey faced by rare diseases patients. The Constituent and Scientific Committees identify and propose actionable topics for Task Forces to tackle, and issue recommendations to drive policy and systemic changes. Obtaining a molecularly‐confirmed diagnosis for rare disease patients is indispensable, in order to assure optimal disease management, lessen complications of the disease, advance genetic counseling, and provide important psychosocial benefits to patients and their families. At present, the causative gene is still unknown for approximately half of all rare diseases and only 15% of rare genetic diseases have an easily available single‐gene diagnostic test in the 40 countries of the Orphanet consortium.29, 30 The Diagnostic and Interdisciplinary Scientific Committees investigated the issues surrounding gene discovery and diagnostics of rare genetic diseases, and developed areas and strategies for improvement, including: the use of ontologies for exchanging clinical data; the development of interoperable tools and resources to facilitate genomic and phenotypic data ascertainment and analyses; the development of best and ethical practices in data discovery and sharing; the use of genetic and functional evidence for gene discovery; and the increased investigation of novel disease mechanisms.4 Every successful gene discovery will not only unlock a potential diagnostic opportunity, but also has the potential to contribute to preventive and therapeutic opportunities for the corresponding rare disease, enabling precision medicine approaches for this patient population. Therefore, such genetic linkages to rare diseases are extremely important and have incredible potential for rare disease patients. A further consideration is that equitable diagnostic, preventative, therapeutic innovation, and global scope also require the generation, detailed understanding, and implementation of normative genomic and phenotypic data, especially for those currently underrepresented in such efforts, including across Europe, Asia, the Americas, and in Australia—Aboriginal and Torres Strait Islanders.31, 32 It is likely that this new knowledge will also unlock insights for currently more well‐studied populations. Therapeutic development is a time‐ and resource‐intense process, and often rare disease patients do not have the luxury to wait nor the means to access therapies, if available. The Therapies Scientific Committee explored the steps along the drug development process, from target identification to drug approval by the regulatory agencies, and developed a set of recommendations that further foster therapeutic development of orphan medicinal products aligned with scientific and regulatory guidance.33 Three key points were emphasized as important to speeding up the process: soliciting protocol assistance from regulators at every stage of product development; enhancing early and continuous dialog with all stakeholders to ensure patients’ needs are addressed; and collaborating with health technology assessment bodies and payers to ensure that therapies brought to market are accessible to patients. The Therapies Scientific Committee is also cognizant of the need to harmonize best practices among the major regulatory agencies in order to bring about a transformational boost in the development of new rare disease therapies. Efforts by regulators to align systems and practices are ongoing and IRDiRC aims to enhance and support such cooperative and collaborative actions between researchers and regulators.19 Task Forces In order to tackle specific topics of rare diseases research, and to further contribute to the IRDiRC objectives, topic‐specific time‐limited Task Forces are established. Each Task Force reviews current impediments to efficient and effective research and proposes solutions through policy recommendations and/or technical applications including platforms, tools, and standards. These Task Forces may operate solely as IRDiRC initiatives, or jointly with partner groups that wish to collaborate and address similar issues. They are also comprised mostly of external expert members to ensure that different perspectives are cultivated to drive innovation and new approaches. Matchmaker Exchange, a federated network of genotype and phenotype databases which allows for rare disease gene discovery,34 is a collaborative action between IRDiRC, the Global Alliance for Genomics and Health (GA4GH),35 and other organizations. IRDiRC also partners with GA4GH on the Automatable Discovery and Access Task Force to develop a standardized way to represent consent and other conditions of clinical data use, making the information computer‐readable and available for automated search and sharing activities36; and on the Privacy‐Preserving Record Linkage Task Force, which was set up to develop guidelines on the ethical, legal, and technical requirements of participant identifiers in rare diseases research, and investigate a technical solution for deduplication of research participants in data sets without knowledge of their identities.37 Other IRDiRC Task Forces include: i) the International Consortium of Human Phenotype Terminologies, which provided the community with standards to achieve interoperability between databases by enabling the linkage of phenotype and genotype databases for rare diseases38; ii) the Patient‐Centered Outcome Measures Task Force, which made recommendations on methods to support the development of patient‐relevant outcome measures for rare diseases in order to improve the quality of future trials and to provide data of relevance to the patient community39; iii) the Small Population Clinical Trials Task Force, which produced recommendations for efficient and innovative clinical trial designs relevant to small populations, often the focus of rare disease clinical trials, using scientific advice from regulators40; and iv) the Data Mining and Repurposing Task Force, which identified opportunities to leverage the existing research and patient data to realize the full potential of data mining and drug repurposing.41 As new issues and areas of improvement are identified, Task Forces are set up, aiming to further contribute to diagnostic, therapeutic, and interdisciplinary development, and optimize the global rare diseases research enterprise. “IRDiRC Recognized Resources” Just like rare disease patients, the researchers who study rare diseases are often few and geographically spread. As such, resources and tools available to further their work are not always known to them. To address this need, IRDiRC set up an indicator called “IRDiRC Recognized Resources” to highlight publicly available resources that researchers in the rare diseases community have found useful and, if were to be used more broadly, may accelerate the pace of rare diseases research.42 To receive this designation, resources are evaluated via a peer‐review process by IRDiRC Scientific Committee members and experts in the field; applications are accepted on a rolling basis. To date, 17 resources have received the label: five guidelines, five platforms, three reference databases, two standards, and an advisory committee. Partner Initiatives IRDiRC successfully collaborates with several partner initiatives to develop global policies and recommendations, build technical solutions for specific rare disease issues, or plan joint projects and events. These initiatives include, but are not limited to, Orphanet,17 GA4GH,35 the Global Genomic Medicine Collaborative (G2MC),43 RD‐Connect,44 TREAT‐NMD Alliance,45 RARE Bestpractices,46 RD‐Action,47 the European Reference Networks (ERNs),48 the US Office of Rare Diseases Research (ORDR) with its Rare Diseases Clinical Research Network (RDCRN) at the NIH's National Center for Advancing Translational Sciences,49 and the Undiagnosed Diseases Network International (UDNI).50 Through these collaborative efforts, there have been marked improvements in disease classification and coding, with a growing acceptance of standard nomenclature and development of policies for ethical and secure data sharing for rare and genetic diseases.51, 52, 53, 54 This is being complemented by significant developments in genomics knowledge and technologies, which are driving faster and more accurate diagnoses25, 55 and the development of specific treatments, labeled as “precision medicine” or “personalized medicine.” While the benefits to individuals of such targeted approaches are clear, the same knowledge and technologies are providing opportunities to better understand and assess the impact of disease at a population level. In line with this, an emerging precision public health paradigm is leading to the development of policies and programs targeted to at‐risk groups, in order to improve the overall health of the population.56 The use of genomics in such public health approaches is key to driving improvements in healthcare delivery for people living with rare diseases. The catalytic drive for this research is patients living with rare diseases and their families. They are the single most transformative force of the rare diseases sector, in areas that include, for example, research and clinical networks, new approaches to therapies and clinical trials, gene and disease pathway discoveries that unlock new knowledge, data sharing and creation of matchmaking platforms, and the translation of this new knowledge into the public health setting for the benefit of all. In partnerships around the globe, patients and patient organizations have joined with governments, industry, academia, regulators, healthcare professionals, and philanthropic organizations to speak with one voice. People living with rare diseases and their families seek a diagnosis that will enable doctors and other professionals to provide the best care in their setting, and improve their journey. To achieve these outcomes and the equitable care citizens expect of their health system, many rare disease patients willingly make public the details of their lives and the lives of their loved ones. This, in and of itself, might seem to be a higher price to pay than others with a smaller health burden are expected to pay. However, the reality is that the rare diseases community has increasingly become self‐organized through social media networks and patient organizations, many of which are international. A number of organizations are truly global and provide voice to more than 350 million people living with rare diseases worldwide. In this article, the authors are not able to do justice to the phenomenal drive and empowerment derived from the national and international patient organizations and networks. As an example, two recent global developments, the launch of the Rare Diseases International (RDI) and the inauguration of the newly formed nongovernmental organization (NGO) Committee for Rare Diseases, established under the umbrella of the Conference of NGOs with Consultative Status to the United Nations Economic and Social Council (CoNGO), serve as a testament to the escalation in the international networks of patient organizations, and also to the many decades of accumulated and expanding influence of rare diseases patients and their families in driving change.57, 58 Interaction with External Stakeholders A major tenet of IRDiRC is the promotion of collaboration, and technology and knowledge sharing. To this end, interactions with external stakeholders are of utmost importance to achieving the IRDiRC objectives. In addition to its many partnered initiatives to develop global policies, IRDiRC recruits expert nonmembers to populate its Scientific Committees and Task Forces. For its Scientific Committees, IRDiRC members identify appropriate expert opinion leaders to ensure diverse geographical representation and breadth and depth of scientific expertise. IRDiRC's expertise, knowledge base, and scientific networks are further broadened by the Task Forces, which are assembled with a very specific purpose, with clear time‐delimited aims and milestones. Through this process IRDiRC members and Scientific Committee members engage and involve domain and subject matter experts throughout the world to achieve maximally informed and impactful results. For example, several Task Forces of the IRDiRC Diagnostics Scientific Committee have partnered with the GA4GH (see section above for details about the Task Forces). IRDiRC members give presentations on IRDiRC in a wide range of international settings, while also reaching out to other organizations, networks, and individuals to join the consortium and contribute their insights to achieving the IRDiRC goals. To advance IRDiRC's impact and reduce unintended duplication, the Scientific Committees and Task Forces regularly publish open‐access articles on their work, complemented by recommendations and reports published on the IRDiRC website and promoted via social media. These dissemination efforts are crucial not only to the translation of new knowledge and insights across the scientific and medical communities, but also to the provision of relevant and authoritative information on the state of the rare disease field to our most valuable partners, the people and families living with rare diseases. CONCLUSION The past 6 years have been remarkable for the rare diseases research community and for rare disease patients. Tremendous progress has been made toward improved diagnostics and patient care. Major public‐sector research initiatives focused in this area have emerged in many countries, and most notably in the US NIH, the EC, and the AMED. However, the further transformative aspect has been the engagement and collaboration between these public funders, industry, and the people living with rare diseases. This being said, substantial challenges remain. Next‐generation genomics and improved data sharing have resulted in faster discovery of causative genes and consequent development of new diagnostics, although there are signs that the rate of disease gene discovery is now slowing as the remaining unsolved diseases are likely more complex. New approaches, including those better interrogating the nonprotein‐coding majority of the genome will need to be developed to address this next step. The rate of rare disease therapeutic development is increasing and there is substantial cause for optimism with regard to innovative therapies. However, the translational rate of these developments to marketing approvals is still relatively low and it remains the case that over 90% of rare diseases lack a specific treatment. Today, it has been estimated that only 6% of rare diseases currently have available treatment, of which less than 1% are curative, leaving a large majority of patients still awaiting a therapy.59, 60 Parallel to the development of innovative therapies, the potential provided by data‐mining and repurposing approaches has not been fully realized. Serious inequities also exist with respect to patient access for those diseases for which an effective treatment is available. In addition, it is not clear that the present model for recovering drug development costs from market sales can be extrapolated to rare diseases with worldwide patient populations of hundreds or less. Novel approaches to therapeutic discovery, as well as new models both for funding drug discovery and for covering treatment costs, are likely to be necessary for rare diseases to be treated in a comprehensive manner. Informed by the phenomenal progress enabled by the Scientific Committees, the Consortium has recently instituted three sector‐specific Constituent Committees (Funders, Companies, and Patient Advocacy Groups), which are identifying areas of common need and opportunity within their sector, and prioritizing and addressing solutions via coordinated implementation activities.20 IRDiRC has been a major positive factor in raising public awareness of rare diseases, the need for more research to address them, and of collaborative tools that allow ethical data sharing for and with patients. It has clearly led to increased investment of public sector research funds for rare diseases. IRDiRC has helped to catalyze several important initiatives that are improving collaboration among researchers and enhancing the ability of patients to engage as constructive partners in research. While considerable headway has been made, there is a need to accelerate progress, building on the willingness and impetus to collaborate across borders, to ensure that IRDiRC's new goals for the rare diseases community for the next 10 years will not only be achieved, but surpassed. This time next year,‐ this time ten years,‐ this time one hundred years……I cannot doubt but that these things, which now seem to us so mysterious, will be no mysteries at all; that the scales will fall from our eyes; that we shall learn to look on things in a different way ‐ when that which is now a difficulty will be the only common sense and intelligible way of looking at the subject. —Sir William Thomson, Baron Kelvin of Largs, 1889 Conflict of Interest The authors declare no conflicts of interest. Supporting information Supporting Material Click here for additional data file.

          Related collections

          Most cited references19

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Future of Rare Diseases Research 2017–2027: An IRDiRC Perspective

          The International Rare Diseases Research Consortium (IRDiRC) was founded in 2011 with the conviction that rare diseases research had reached a critical juncture. Proof of principle existed that rare diseases could be diagnosed, new treatments successfully developed and approved, and improvements in quality and quantity of life achieved. Government research funders, companies, scientists, and patient advocacy groups had all demonstrated their commitment and effectiveness in contributing to progress in rare diseases research. However, the work was largely atomized, with each organization, each country, and the champions of each disease pursuing independent, often duplicative solutions. The scale of the “rare disease problem”—thousands of rare diseases, the vast preponderance of them with no approved treatment, and decades‐long diagnostic odysseys for many patients—led to the realization that the time had arrived for global cooperation and collaboration among the many stakeholders active in rare diseases research, to capitalize on these proofs of principle, and maximize the output of rare diseases research efforts around the world. IRDiRC's initial aims were to aid in the achievement of two overarching objectives: to contribute to the development of 200 new therapies and the means to diagnose most rare diseases by the year 2020.1 For more detailed information on the history, governance, and nascent stages of the Consortium, please refer to the accompanying piece on the first 6 years of IRDiRC.2 Due to the remarkable global surge in activity in rare diseases research over the last 6 years, including contributions by IRDiRC, the Consortium's 2020 goal for 200 new therapies was achieved in early 2017—3 years ahead of schedule—and the goal for diagnostics—the ability to diagnose most rare diseases by 2020—is within reach; these accomplishments were celebrated at the 3rd IRDiRC Conference in Paris in February 2017.3 The 6 years preceding this 2017 conference have been truly extraordinary for the rare diseases research community and for rare disease patients. Major public‐sector research initiatives focused in this area have emerged or expanded in many countries, most notably from the US National Institutes of Health (NIH), the European Commission (EC), and the newly formed Japan Agency for Medical Research and Development (AMED). Engagement and partnering among public funders, scientists, industry, and people living with rare diseases have gone from being the exception to commonplace. IRDiRC has been a major positive factor in raising public awareness about rare diseases, the need for more research to address them, and for collaborative tools which allow ethical data sharing for and with patients. It has also clearly led to increased investment of public‐ and private‐sector research funds for rare diseases, in addition to the research funding raised by patients and patient organizations. IRDiRC has helped to catalyze several important initiatives that are improving collaboration among researchers and enhancing the ability of patients to engage as constructive partners in research.2, 4 As gratifying as these developments are, those who lead much of the global rare diseases research community are well aware of the enormous challenges that lie ahead for all patients living with rare diseases to receive an accurate and timely diagnosis, to have approved treatments available, to get access to those treatments, and to realize improvements in their quality and quantity of life; in short, to be able to live the best life possible. Although the means to diagnose most rare diseases that are caused by mutations in the coding genome is on track to be achieved either via genotype–phenotype correlation or novel gene discovery, in practice most patients with rare diseases spend years in the healthcare system before an accurate diagnosis is made. For rare diseases that have yet to be defined, next‐generation genomics and improved data sharing have resulted in faster discovery of disease gene and consequent development of new diagnostics, although there are signs that the rate of disease gene discovery is now slowing, as the remaining unsolved diseases are likely more complex.5 To respond to this subsequent level of complexity, novel approaches, particularly ones that better address the nonprotein coding regions of the genome, will need to be developed. There is some cause for confidence with regard to new therapies as the rate of rare disease therapeutic development has been increasing. However, it remains the case that 94% of rare diseases lack an approved treatment,6, 7 that the number of currently untreatable rare diseases that have a first treatment approved each year remains low, and that serious inequities remain with regard to patient access to effective treatments even when they are available. Additionally, the present model for recovering drug development costs from market sales has not been proven in its application to rare diseases with worldwide populations of hundreds or less. To address these therapeutic discovery issues, new approaches including data‐mining and repurposing, in addition to new models for funding drug discovery and covering treatment costs, will be necessary for the comprehensive treatment of rare diseases worldwide. With this paradox in mind—the desire to celebrate unprecedented progress but recognizing the immense need and opportunities that remain—IRDiRC set about devising new global rare disease goals for the coming decade. Through this year‐long collaborative process, IRDiRC aimed to set goals that would achieve all that is scientifically possible in the short term, and aggressively push the limits of what is currently impossible in the longer term, all with the knowledge that patients are waiting, and “Time Equals Lives.”8 IRDiRC: 2017–2027 Process To assure input from all stakeholders and arrive at a short list of ambitious but achievable IRDiRC goals for the next decade, a multistep, year‐long, objectives‐setting process was implemented. Initially, ideas on critical problems in the rare diseases field and solutions to them were solicited broadly from IRDiRC member organizations and nonmembers represented on the IRDiRC Scientific Committees from academia, patient organizations, the biopharmaceutical industry, and regulatory bodies, in each of the three scientific focus areas: diagnostics, foundational/interdisciplinary, and therapies. The hundreds of ideas submitted were grouped and consolidated, debated online and in‐person, and then voted on to determine which were of highest priority and need. Based on this process, a series of potential goals were generated, along with activities to advance the goals, and metrics to measure progress. During the internal IRDiRC Meeting in Paris, France in February 2017, the candidate goals, activities, and metrics were further refined by the IRDiRC members. At the open IRDiRC Conference that followed, the goals were presented to the greater rare disease community for feedback, discussion, and questions to further shape the IRDiRC vision and objectives for the next decade. This vigorous, animated, and informed session added broad public input to the goal‐setting process and in addition spurred excitement and engagement about IRDiRC's plans to deliver on the promise of science for people living with rare diseases over the next decade. Following the Meeting and Conference, the final IRDiRC Vision and 2017–2027 Goals were formally adopted by vote of the IRDiRC Consortium Assembly. Framework Given the unusually broad scope of IRDiRC—in science, constituencies, and geography—the IRDiRC goal‐setting process incorporated an unusually broad series of criteria. First, the process utilized the “SMART” criteria—that is, candidate goals needed to be Specific, Measureable, Achievable, Realistic, and Timely. They also needed to be within the scope of IRDiRC's research mission. Lastly, they needed to be easily understood by a wide variety of stakeholders and audiences, while also being bold and transformational. Therefore, it was determined to organize the process on four levels—Vision, Goals, Activities, and Metrics. The Vision is IRDiRC's overarching statement of the end state toward which all its activities drive; the Vision is aspirational and not time‐delimited. The Goals state bold but distinct achievements that IRDiRC commits to accomplish over the next 10 years that will advance the realization of the Vision. The Activities are discrete, shorter‐term projects IRDiRC will perform to advance each of the Goals; the list of Activities will continually change, depending on successes and failures of previous Activities and evolution of the field. The Metrics will assess and track progress of Activities over time, ensuring accountability and progress toward the Goals. The new IRDiRC Vision, the IRDiRC Goals for 2017–2027, and exemplar Activities and Metrics, follow. The IRDiRC Vision Enable all people living with a rare disease to receive an accurate diagnosis, care, and available therapy within 1 year of coming to medical attention IRDiRC is well aware of the aspirational nature of this Vision; IRDiRC is also cognizant that some aspects of the Vision are outside its research mission. However, IRDiRC also believes that the Vision is achievable with all stakeholders’ commitment, cooperation, and collaboration. Thus, the challenge inherent in the Vision is intentional, aimed at galvanizing the broad rare disease community, within IRDiRC and outside it, to not only enable universal diagnosis and treatment, but also ensure that these interventions reach people with rare diseases, and have the intended positive impact on their health and well‐being. The IRDiRC Goals for 2017–2027 The three Goals that IRDiRC members have committed to achieving in the next decade to advance the realization of the IRDiRC Vision follow, along with the rationale, challenges, and opportunities of each. Goal 1: All patients coming to medical attention with a suspected rare disease will be diagnosed within 1 year if their disorder is known in the medical literature; all currently undiagnosable individuals will enter a globally coordinated diagnostic and research pipeline Recent data indicate that approximately half of the individuals with a suspected rare disease are undiagnosed, while those who have received a diagnosis wait on average 5–6 years,9, 10 and diagnostic delays of several decades have been observed. The clinical introduction of new diagnostic methods such as next‐generation sequencing has allowed the laboratory turnaround time to be as short as several weeks to diagnose some of the rare diseases with a known molecular basis. Each undiagnosed rare disease represents an opportunity to open up a new area of biological insight, and it follows that, as the number of novel genes and pathogenic variants identified increases, so does the diagnostic yield. The time has come for researchers, clinicians, and patients worldwide to collectively understand the etiology of the vast number of rare diseases, make the final push to enable the diagnosis of all rare diseases, and facilitate access to an efficient diagnosis for patients. Within the next decade, IRDiRC will work together to implement a system whereby patients with a suspected rare disease of known molecular basis will be diagnosed within 1 year of initial presentation to a medical professional instead of confronting a years‐long diagnostic odyssey. The challenges in achieving this part of Goal 1 are principally operational, involving public and physician awareness, efficient referral within the medical system, and the requirement for radically more efficient sharing of diagnostic expertise and data among practitioners and researchers worldwide. Rare disease mechanism discovery The number of unsolved patients following whole exomes sequencing argues that more disease genes and variants await discovery, thus the discovery effort must be expedited. So far, most of the known disease variants fall in coding regions of the genome, but much less is known, for example, about the role of noncoding region variants and structural variants in disease.5 This calls for approaches that are complementary to whole exome sequencing (WES) such as whole genome sequencing (WGS), long read technologies, and transcriptome sequencing that can more effectively target noncoding regions and/or structural variants. Moreover, variant interpretation still needs improvement through developments in bioinformatics, analysis algorithms, and data sharing. Wide acceptance of data standards and ontologies (e.g., Human Phenotype Ontology (HPO)11 and Orphanet Rare Disease Ontology (ORDO)),12 and automated exchange of phenotypic and genomic information via shared platforms and tools (e.g., Matchmaker Exchange13 and RD‐Connect14) should be required to transform sequence information into diagnostic knowledge. Functional analyses at scale will need to be developed to facilitate variant interpretation in conjunction with data sharing. Patient access to diagnosis After initial presentation to a medical professional, patients with a rare disease often spend a long time trying to find a specialist with appropriate expertise to recognize the syndrome or perform the correct diagnostic test. Comprehensive and easily accessible information about subspecialty medical professionals and diagnostic laboratories can help shorten this time. In conjunction, sequencing and analysis costs will need to continue to drop to improve affordability. Finally, robust data should be collected and analyzed on diagnostic utility, clinical utility, and cost‐effectiveness to facilitate reimbursement of sequencing‐based diagnosis by more health insurance companies. International network for undiagnosed patients It has been shown that undiagnosed patients have an increased chance for their diagnostic challenge to be “solved” in a research setting where more comprehensive sequencing, analysis, and data sharing can be performed. It is time to establish global networks of clinical and research laboratories to collectively tackle undiagnosed diseases. Ideally, appropriate consents including the provision for research and data sharing should be obtained from the outset of the clinical testing process. Samples for further research using sequencing and other genomic methods should be collected and stored in appropriate biorepositories. However, there is only so much that can be done without cooperation and coordination on a larger scale. If a diagnosis is not made after initial sequencing, then the data should be immediately transferred to a global network of appropriate expertise that can accept it for further study and immediate feedback of the result. The Undiagnosed Diseases Network International (UDNI)15—modeled after the US NIH's Undiagnosed Diseases Program (UDP)—is an example of a program established to aid in this effort. Collaboration with the UDNI as well as UDP will bring crucial attention to complex cases, where collective expertise will lead to a higher chance of providing a much‐needed diagnosis in order to identify the best course of treatment for each patient. IRDiRC also encourages collaboration with national programs, such as the Japanese Initiative on Rare and Undiagnosed Diseases (IRUD),16 to capitalize on additional knowledge and data sharing with the aim of bringing diagnoses to rare diseases patients. Education of physicians and engagement of patients To take advantage of fast‐evolving technologies, established networks, and available tools, it is necessary to educate physicians and engage patients and families. For example, education can be provided via courses on rare diseases and new diagnostic methods and targeted to various end‐users, including physicians and patients, with different levels of knowledge at the outset. Patient engagement in research and clinical networks should continue to be facilitated. Metrics Online Mendelian Inheritance in Man (OMIM)17 and Orphanet18 will continue to be reliable resources for monitoring newly reported diseases and disease genes. The time it takes for a patient to be diagnosed could come from surveys of specialty physicians, clinical labs, and patient organizations, or more targeted sampling via rare disease networks. Goal 2: 1000 new therapies for rare diseases will be approved, the majority of which will focus on diseases without approved options Although the rate of therapy development for rare diseases has been increasing, the fact remains that most rare diseases—well over 90%—lack an approved treatment and the number of currently untreatable rare diseases to receive a first treatment each year remains low. The introduction of regulations, policies, and incentives dedicated to orphan drug development has spurred significant investment in therapeutic development to the benefit of rare disease patients.19 Since 2010, IRDiRC has tracked the number of orphan medicinal products (OMPs) receiving first approval for a new indication in the European Union and/or the United States, and has found an increase from 15 in 2010 to more than 40 in 2014 and 2015, with a current average of ∼35 approvals per year. Between 2010 and 2016, over 220 OMPs have received first approval for a new indication in the European Union and/or the United States. While significant, this achievement does not negate the fact that patients with one of the thousands of other rare diseases are still waiting for a therapy to be approved for their conditions. Innovative approaches, including clinical trial design, data and specimen collection, clinical end points, repurposing, natural history studies, and engaging the many players involved are necessary for exponentially improving therapy development on a global scale. Therapeutic development pipeline Assuming a constant delivery of OMPs from the pipelines of biopharmaceutical industries, in the next 10 years treatments would become available for only ∼600 of the 7,000 known rare diseases. Thus, new approaches will be needed, particularly since the current pace may not be sustained. In 2016, only 34 new indications were approved, suggesting a slowing of the development and approval pace. Moreover, developed drugs have often clustered around similar technologies or therapeutic approaches that will soon have maximized their capacity to generate new therapeutic advances. For instance, the systemic manifestations of several lysosomal storage disorders (LSDs) have been quite successfully addressed by treatments based on enzymatic replacement through recombinant proteins containing mannose‐6‐phospate residues or small molecules through substrate inhibition, but the list of remaining LSDs to be targeted is significantly shrinking and all these drugs leave unaddressed the same manifestations (e.g., the central nervous system involvement). The “lower‐hanging fruits” of easily developable indications, addressable by traditional approaches, will likely decrease over time, leaving more complex therapeutic targets and yet unproven technologies. In addition to challenges specific to rare diseases, the risk‐adjusted development costs in the pharmaceutical industry have witnessed an overall increase, and postregulatory approval access challenges have become larger due to budgetary constraints of payers.20, 21 In order to achieve the IRDiRC goal of 1,000 new therapies in the next decade, a significant increase in R&D productivity is needed, with a compounded annual growth rate at or above 10%, thus tripling the current rate. Moreover, the IRDiRC goal is for new orphan drug approvals to be predominantly for diseases currently without approved drugs. Although IRDiRC anticipates that many of the 1,000 new approvals will be new indications for existing agents rather than new molecular entities, scalability and sustainability will be significant challenges, both to the regulatory system, and to healthcare budgets. IRDiRC includes representatives from the world's major pharmaceutical regulatory agencies and is deliberately increasing representation from health technology assessment agencies, in order to anticipate and mitigate these challenges. Potential advancements in therapeutic development This important goal can be achieved only through a dramatically more efficient development process driven by a radically new approach utilizing common standards across distinct research fields, sharing of best practices, creating sustainable business models, and redefining the regulatory environment. New methodologies are needed to streamline drug development. These include early stage improvements such as increasing the efficiency of data collection and sharing, improving the understanding of disease progression and phenotypes, improved methods for preclinical assessment of safety and efficacy, and methodologies for small size clinical trials. In addition, later stage advancements including defining end points more universally suitable for measuring patient's benefit, providing medical relevance, generating regulatory benefit/risk evidence, and quantifying a product's economic value for payers, companies, and society at large are essential. The emerging European Reference Networks22 and the potential collaborations with the US Rare Diseases Clinical Research Network23 provide an unprecedented opportunity for coordinating global rare diseases research to: improve care standards, increase access to diagnosis and treatment, increase the understanding of phenotypes and natural history, increase enrollment of patients into clinical trials, and more effectively create and manage disease registries. Engaging patients and regulators Placing patients at the center of clinical research, drug development, and evaluation is increasingly recognized as paramount to fully understanding a disease and to identifying meaningful end points. Their knowledge, contribution, empowerment, and participation are crucial to increasing the efficiency of such efforts. Close cooperative actions with regulators will also be indispensable, particularly via early dialog with regulators and product development with protocol assistance to ensure regulations are adhered to at every step, thus maximizing the potential outcome of a marketing authorization. To cope with an increased volume of applications and requests of protocol assistance, a number of changes will be necessary: streamlining the approval process, creating collaborative review processes between regulators from different jurisdictions, increasing human resource and training programs, and potentially updating regulations to assist in accelerating therapy development. Efforts are already under way to streamline and align regulatory processes across jurisdictions24; IRDiRC aims to aid and foster such efforts, as they will ultimately contribute to the development of new rare disease therapies. In addition to coordinating research efforts, data sharing, and patient engagement, it is also vital to promote changes to the drug development landscape such as new models of risk and incentive sharing between public and private partners, systematic repurposing of existing agents, and developing a more flexible regulatory framework. IRDiRC is committed to work as a key enabler of this quantum change as reflected in the vision. IRDiRC promotes the development and sharing of new tools, best practices, and recommendations to inform research policies and strategies worldwide. IRDiRC also will foster new methods to enable dialog between private and public research funders and regulators with the goal of bringing about this quantum change needed to reach the ambitious goal of developing 1,000 new therapies within the next decade. Metrics The number of new indications treated with medicinal products for rare diseases receiving marketing authorization in the European Union, the United States, and Japan will be the main indicator of progress toward the 1,000 therapies goal, based on information from the European Medicines Agency (EMA), the US Food and Drug Administration (FDA), and the Japanese Pharmaceuticals and Medical Devices Agency (PMDA). A number of secondary metrics will also be developed to monitor the quality of evolution of the field, e.g., the number of medicinal products for rare diseases with marketing authorization but without orphan designation, and the number of RDs that are addressed by these medicinal products. Goal 3: Methodologies will be developed to assess the impact of diagnoses and therapies on rare disease patients While faster diagnosis and increased development of new therapies are essential, their impact on people living with rare diseases cannot be assumed; for example, patients can benefit only to the degree that they have access to the interventions, and access may or may not lead to the intended improvement in quantity and/or quality of life. Although IRDiRC members, representing funders, companies, patient advocacy groups, scientists, and other stakeholders agreed on this truism, they varied in their view of IRDiRC's role, as a research organization, in addressing the impact issue. While patient advocacy group members tended to support inclusion of impact assessment as critical IRDiRC research, many scientific and funder members felt that this was more the mandate of health technology assessment authorities. A rich and important debate, including with numerous stakeholders participating in the IRDiRC Conference in February 2017, concluded with the realization that no matter what organization is charged with impact assessment, the methods to do that assessment are currently woefully inadequate, and that IRDiRC therefore could and should focus on the development of improved methodologies and tools for performing such impact assessments. Appropriate access to diagnosis and treatment depends on a multitude of factors, including clinical guidelines and recommendations; regulatory policies; pricing; insurance, coverage, formulary, and reimbursement; and even the awareness of healthcare providers. The efficiency and extent of translation of diagnostic and treatment developments into tangible outcomes and practice are currently hampered by limited assessment of their impact on patients. Development of robust methods to measure access, effectiveness in real‐world settings, and impact on patient outcomes will therefore be a focus of the IRDiRC over the next decade. Such research needs to particularly involve underdeveloped areas worldwide, which starts by expanding our global footprint into more underrepresented regions and the inclusion of such members in all activities. Equally important, as a global health issue, IRDiRC members are committed to implement these advances equitably to reduce existing and potential health disparities. Such disparities include those between Indigenous and non‐Indigenous peoples, which, at its core, requires indigenous‐specific reference genetic data sets to improve clinical diagnosis and optimize therapies.25, 26, 27, 28 We anticipate that this research will not only benefit rare disease patients worldwide, but also have impacts in the wider context of personalized medicine. Measurement of impact Assessing the impact of diagnosis remains a complex issue. Counting the number of diagnostic tests might be relatively simple given the various worldwide, country‐specific, and company‐specific listings but is nonspecific and indirect, speaking only to availability and not access. Quantifying the number of people who have received a diagnosis, the length of the diagnostic odyssey, and the impediments to diagnosis is, however, not straightforward. This quest goes beyond the mandate of a single clinical or research team. Aggregating this information requires a multidisciplinary and multistakeholder approach that must navigate the continuum from clinical research to healthcare services in multiple systems and cultures. Thus, measuring the impact of diagnosis may include such items as quantitative and statistical analysis, assessment of quality of life, and/or economic dissection of repercussions on medical care. Assessing the impact of treatment is also largely limited to regulated therapies, since these are easier to count. It has been suggested that other types of treatments or interventions, such as nonpharmaceutical approaches, physical and behavioral therapies, and/or devices may be as valuable to patients as “drugs,” but these are generally neglected when considering impact. Similarly, research into healthcare system optimization and the implementation of recommendations to improve its functions may have an important impact on patients’ outcomes and health. The use of existing tools and platforms, e.g., the NIH Genetic Testing Registry,29 and RARE‐Bestpractices,30 that help develop rigorous process and qualitative markers for the evaluation of the diagnostic and modes of care should be factored into any methodology development. The funders should engage in identification and financial support of research projects that will tackle the complexities around the measurement of health outcomes. Research in health systems, economics, and ethical frameworks should also be promoted. Furthermore, IRDiRC may consider how to engage appropriate stakeholders in healthcare systems to ensure that any methods developed could be recognized rapidly, and applied at both the national and international levels. CONCLUSION The members of IRDiRC and, more important, their organizations, have committed themselves to an ambitious set of 10‐year goals that will advance the realization of the IRDiRC vision of prompt and accurate diagnosis, effective treatment, and amelioration of illness for all people living with rare diseases. In an ambitious and multifaceted project like IRDiRC's, coordination and monitoring of progress will be essential; these will be performed by the IRDiRC Committees and Scientific Secretariat. The Committees and their Task Forces will promote activities to advance the goals, and metrics will be applied to monitor progress toward the goals. Some may question whether the costs of IRDiRC's vision can be justified, given the low prevalence of these disorders. We, the leadership of IRDiRC, believe that, to the contrary, the global community cannot afford not to achieve these goals, and the resources the member organizations are committing to rare diseases research to realize the IRDiRC vision testify to this conviction. On a purely financial level, the cost to health systems of caring for people whose rare diseases are undiagnosed or untreatable are disproportionate and growing. On a human level, we believe that every person with an illness, whether rare or common, has the same right to a diagnosis and treatment, and that the contributions to humanity of rare disease patients are well beyond our imagining. We are all familiar with how people living with HIV/AIDS, once an undiagnosable, untreatable rare disease, are continuing to enrich the human family in innumerable ways, and how much the work to diagnose and treat HIV/AIDS taught us about human biology and other diseases. This is our vision for the millions of people living with the thousands of other rare diseases. As leaders of the global rare diseases community in the public and private sectors, we are under no illusions about the challenges to achieving our new goals by 2027. However, we are equally aware of the epochal advances in rare diseases science and medicine over the last decades, the evolution of a culture of collaboration, teamwork, and common cause that now unites the rare disease community, and the reality that the pace of progress is positioned to accelerate. These are goals that can only be achieved with fundamental changes in the conduct and sharing of science, and application of that science as rapidly as possible to advance the care of rare disease patients—changes IRDiRC members have committed to catalyze. We believe that these goals are eminently achievable over the next decade—but only with continued commitment to scientific excellence, rapid and ubiquitous sharing of approaches and data and resources, and continued monitoring of progress and constant reevaluation of direction based on new data. IRDiRC's is a rigorous, noble—and achievable—vision, which we believe will bring out the best in science, in medicine, and in ourselves. We welcome new members, who share our vision and commitment to action, to join us. And we look forward to updating the community on our progress. Conflict of Interest The authors declared no conflicts of interest. Supporting information Supporting Information Click here for additional data file.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Rare diseases, orphan drugs and their regulation: questions and misconceptions.

            Sustained advocacy efforts driven by patients' organizations to make rare diseases a health priority have led to regulatory and economic incentives for industry to develop drugs for these diseases, known as orphan drugs. These incentives, enacted in regulations first introduced in the United States in 1983 and later in Japan, Europe and elsewhere, have resulted in substantial improvements in the treatment for patients with a range of rare diseases. However, the advent of orphan drug development has also triggered several questions, from the definition of rarity to the pricing of orphan drugs and their impact on health-care systems. This article provides an industry perspective on some of the common questions and misconceptions related to orphan drug development and its regulation, with the aim of facilitating future progress in the field.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              ‘You should at least ask’. The expectations, hopes and fears of rare disease patients on large-scale data and biomaterial sharing for genomics research

              Within the myriad articles about participants' opinions of genomics research, the views of a distinct group - people with a rare disease (RD) - are unknown. It is important to understand if their opinions differ from the general public by dint of having a rare disease and vulnerabilities inherent in this. Here we document RD patients' attitudes to participation in genomics research, particularly around large-scale, international data and biosample sharing. This work is unique in exploring the views of people with a range of rare disorders from many different countries. The authors work within an international, multidisciplinary consortium, RD-Connect, which has developed an integrated platform connecting databases, registries, biobanks and clinical bioinformatics for RD research. Focus groups were conducted with 52 RD patients from 16 countries. Using a scenario-based approach, participants were encouraged to raise topics relevant to their own experiences, rather than these being determined by the researcher. Issues include wide data sharing, and consent for new uses of historic samples and for children. Focus group members are positively disposed towards research and towards allowing data and biosamples to be shared internationally. Expressions of trust and attitudes to risk are often affected by the nature of the RD which they have experience of, as well as regulatory and cultural practices in their home country. Participants are concerned about data security and misuse. There is an acute recognition of the vulnerability inherent in having a RD and the possibility that open knowledge of this could lead to discrimination.
                Bookmark

                Author and article information

                Contributors
                austinc@mail.nih.gov
                Journal
                Clin Transl Sci
                Clin Transl Sci
                10.1111/(ISSN)1752-8062
                CTS
                Clinical and Translational Science
                John Wiley and Sons Inc. (Hoboken )
                1752-8054
                1752-8062
                23 October 2017
                January 2018
                : 11
                : 1 ( doiID: 10.1111/cts.2018.11.issue-1 )
                : 11-20
                Affiliations
                [ 1 ] Office of Population Health Genomics Public Health Division Department of Health Government of Western Australia Perth Australia
                [ 2 ] Directorate General for Research and Innovation (DG RTD) European Commission Brussels Belgium (until April 2017)
                [ 3 ] Merck & Co. Inc. Upper Gwynedd Pennsylvania USA (from June 2017)
                [ 4 ] Department of Biology McGill University Montréal Canada
                [ 5 ] IRDiRC Scientific Secretariat Inserm‐US14, Rare Diseases Platform Paris France
                [ 6 ] National Center for Advancing Translational Sciences (NCATS) National Institutes of Health (NIH) Bethesda Maryland USA
                [ 7 ] Orphanet Inserm‐US14, Rare Diseases Platform Paris France
                [ 8 ] Children's Hospital of Eastern Ontario Research Institute University of Ottawa Ottawa Canada
                [ 9 ] Genetic Services of Western Australia King Edward Memorial Hospital Perth Australia
                [ 10 ] Western Australian Register of Developmental Anomalies Perth Australia
                [ 11 ] Institute of Genetic Medicine Newcastle University Newcastle upon Tyne UK
                [ 12 ] EURORDIS‐Rare Diseases Europe Paris France
                Author notes
                [*] [* ]Correspondence: Christopher P. Austin ( austinc@ 123456mail.nih.gov )
                Article
                CTS12501
                10.1111/cts.12501
                5759730
                28796411
                515a9f55-1a3b-41a2-87ed-5ff59caf2a2b
                © 2017 The Authors. Clinical and Translational Science published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics

                This is an open access article under the terms of the Creative Commons Attribution‐NonCommercial‐NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.

                History
                : 15 July 2017
                : 04 August 2017
                Page count
                Figures: 7, Tables: 0, Pages: 10, Words: 7359
                Funding
                Funded by: SUPPORT‐IRDiRC
                Award ID: 305207
                Funded by: RD‐Connect
                Award ID: 305444
                Categories
                Review
                Research
                Reviews
                Custom metadata
                2.0
                cts12501
                January 2018
                Converter:WILEY_ML3GV2_TO_NLMPMC version:5.2.8 mode:remove_FC converted:09.01.2018

                Medicine
                Medicine

                Comments

                Comment on this article