NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.
Gliklich RE, Dreyer NA, Leavy MB, editors. Registries for Evaluating Patient Outcomes: A User's Guide [Internet]. 3rd edition. Rockville (MD): Agency for Healthcare Research and Quality (US); 2014 Apr.
Registries for Evaluating Patient Outcomes: A User's Guide [Internet]. 3rd edition.
Show details1. Introduction
Once a drug or device is approved for use by a regulatory authority the product is generally used by larger and more diverse populations than are typically studied in the clinical trials leading up to approval. As a result, the period after approval is an important phase for identifying and understanding product safety concerns associated with both acute and chronic use. The need for postapproval (also called postmarketing) safety assessment as it exists today was, for the most part, born out of well-publicized product safety issues that were initially detected by clinicians recognizing a pattern of rare serious events, such as phocomelia caused by prenatal exposure to thalidomide1 and rare vaginal cancers that occurred in young women who had in utero exposure to diethylstilbestrol.2 The detection of serious adverse drug reactions after authorization has led to much debate about the adequacy of both industry and regulatory approaches to preauthorization assessment and testing. However, the decision to authorize a medicine is a balance between wanting to know as much as possible about the safety of a product and the need to make new drugs available for patients.3 The implication is that authorization cannot mean that a medicine is completely safe; rather, it is an assessment that at the time of authorization, the known benefits for the average patient in the approved indication outweigh the known risks. But the degree to which the known risks represent the actual safety profile of a product will depend upon the size, duration, representativeness, and thoroughness of the clinical trial program, which, in turn, is related to the complexity of the patients and the state of knowledge of the disease being targeted. Trials conducted as part of clinical development are, by necessity, of limited duration and size and generally focus on a narrowly defined population that represents only a small segment of the population with the disease or product use of interest. Clinical trial populations tend to be restricted to those who have limited concurrent disease and who are on few, if any, concomitant medications. Typically, trial protocols include lengthy lists of inclusion and exclusion criteria that further restrict the trial population. Unless a drug or a product is intended for a very narrow indication or a very rare disease, it is not feasible to require clinical trials to be inclusive of all types of patients likely to ever be exposed to it. Even in the case of a narrow indication, the potential long-term and delayed effects of a product are unlikely to be established during most clinical trial development programs.
To address the acknowledged limitations of what is known about the safety profile of a product at the time of authorization, postmarketing pharmaco- and medical device vigilance is traditionally, and by regulation, performed through spontaneous adverse event (AE) reporting. The exact requirements for spontaneous reporting to the regulatory authorities vary internationally and depend on the country/region, approval type, and product type. It is widely acknowledged, however, that spontaneous reporting captures an extremely small percentage of the actual events occurring, and that, while it is useful for identifying rare and potentially significant events,4, 5 it has limited use in the detection of other equally important types of events, including increases in events with a high background rate. This form of postmarketing surveillance is reactive in that one waits for AEs/reactions to be spontaneously reported assesses them for causality, and estimates the importance of the information.
As well as collecting only an indeterminate fraction of adverse reactions, this method of surveillance depends upon someone reporting the events of interest. There is some evidence that clinicians who report AEs are not typical of clinicians in general, and other reporters such as patients, lawyers, and consumer groups may have unclear motivations for reporting, which introduces further bias into the equation.6-8
The current methods available for AE reporting are seen by many as burdensome and not amenable to incorporation into a clinician's normal workflow. Waiting for reports to arrive and accumulate may also delay the detection of adverse reactions. On the other hand a massive uptake of a new drug or device, such as seen with Viagra® (sildenafil citrate) or coronary artery stents, may lead to a sudden flood of reports of nonserious as well as serious AEs that could potentially overwhelm established systems.
To overcome some of the difficulties associated with managing large databases of spontaneous AEs, many investigators employ statistical methods to identify signals of disproportionate reporting (SDRs). These methods identify AEs that are reported more frequently with a drug or device than would be expected compared with other event/product pairs in the database, and do not imply any kind of causal relationship.9 It is important to be precise as to what is meant when using the term “signal” or “signal detection” since the terms are ambiguous; in the context of automated methods of detecting statistical anomalies, the term “SDR” should be used.9 However, these statistical methods may not be reliable in certain situations, such as when there is major confounding or when the increased risk is small compared with the background incidence of the event.9 All these above-mentioned limitations mean that there are situations when spontaneous reporting may not be adequate as the sole method of postmarketing surveillance.
To address problems with traditional pharmaco- or medical device vigilance when there are particular known limitations of knowledge of the safety profile of a product and/or to further address unresolved safety concerns, some products are approved subject to postmarketing commitments, which may be requested for safety purposes as well as to address other outstanding questions. In Europe, in response to concerns over pharmacovigilance, marketing authorization applicants are required to submit a European Union-risk management plan (EU-RMP) when seeking a marketing authorization for the majority of new chemical entities and biologics. This EU-RMP states what is known and not known about the safety profile of a medicinal product, how its safety profile will be monitored investigated and characterized and what risk minimization activities will be undertaken. While many products will require only routine pharmacovigilance, for others more proactive methods of pharmacovigilance will be necessary to supplement the use of spontaneous adverse reaction reporting and periodic safety update reports. Although additional clinical trials may occasionally be mandated it is more common for observational pharmacoepidemiological studies to be conducted to ascertain the safety profile of a product under real-world use.
Other observational methods of tracking and evaluating safety data have historically included active surveillance systems, such as the prescription event monitoring (PEM) systems used in the United Kingdom (Drug Safety Research Unit),10 New Zealand (NZ Intensive Monitoring Programme), Japan (J-PEM), and elsewhere, targeting new products; and the retrospective use of administrative claims data. In the United Kingdom, the requirement that access to most secondary care is through a general practitioner has led to the use of their electronic health care systems for pharmacovigilance purposes; however, this type of integrated approach is not yet widely accessible elsewhere. In May 2008, the U.S. Food and Drug Administration (FDA) launched the Sentinel Initiative, an effort to create an integrated electronic system in the United States for AE monitoring, incorporating multiple existing data sources including claims data and electronic medical record systems.11
Medical devices in the United States have surveillance programs different from those for drugs. The Safe Medical Devices Act of 1990 requires that high-risk medical devices be tracked after marketing, and that product corrections and removals be reported to FDA if actions were taken to reduce health risks. Most medical device safety tracking is accomplished through reports submitted to FDA from medical facilities when devices are implanted or explanted. In addition, hospitals, nursing homes, ambulatory surgery centers, and outpatient treatment facilities are required to report to FDA whenever they believe that a device caused or contributed to the death of a patient, though this reporting is a voluntary requirement and not enforceable or audited.12
Whether to comply with a postmarketing requirement or out of a desire to supplement spontaneous reporting, prospective product and disease registries are also increasingly being considered as a resource for examining unresolved safety issues and/or as a tool for proactive risk assessment in the postapproval setting. The advantage of registries is that their observational and inclusive design may allow for surveillance of a diverse patient population that can include sensitive subgroups and other groups not typically included in initial clinical trials, such as pregnant women, minorities, older patients, children, or patients with multiple comorbidities, as well as those taking concomitant medications. In contrast to clinical trials, in which the inclusion criteria are generally tightly focused and restrictive by design, registry populations are generally more representative of the population actually using a product or undergoing a procedure, since the inclusion criteria are usually broad and may potentially include all patients exposed regardless of age, comorbidities, or concurrent treatments. Data collection may lead to insights about provider prescribing practices or off-label use and information regarding the potential for studying new indications within the expanded patient population. Followup duration can be long to encompass delayed risks, consequences of long-term use, and/or effects of various combinations and sequencing of treatments. Such information can be used as a source of publications, to assist the medical community with developing recommendations for monitoring patient safety and product usage, and/or to contribute to the understanding of the natural history of the disease.
There are also many challenges to the utility of registry data for providing more clarity about safety concerns and for prospective risk surveillance. These challenges relate largely to how products are used and the legal, regulatory, and ethical responsibilities of registry sponsors. Most registries that follow specific products do so through cooperation from physicians who prescribe (or implant) these products. Depending on the setup and legal constraints of the registry, sometimes only a subsection of prescribing physicians may be involved in entering patients, a situation that raises questions about the representativeness of the physicians and their patients. However, the registry approach has the potential to be very useful for studying products that are used according to their labeled indications; it also allows for effective surveillance of products that are used off label but by the same practitioners who would use it for the labeled indication. For example, a product might be approved for people with moderate to severe asthma and used off label in patients with mild asthma, yet the prescribing medical providers would already be included in the registry and could easily provide information about all their product use. Off-label use is much more difficult to study when a medical product is used by a wide variety of medical care providers; for example, drugs that promote wakefulness or are thought to increase a patient's ability to concentrate, acting as immunomodulators. The legal, regulatory, and ethical aspects of registry sponsors also affect whether they are required to report any AEs that may be observed since only those legal entities that market (or distribute) a medical product are required to report AEs. For all other parties, such reporting is ethical and desirable, but not enforceable or required.
The purpose of this chapter is to examine the role of registries as one of the available tools for enhanced understanding of product safety through AE detection and evaluation. The examination will include both the role of registries created specifically for the purposes of safety assessment and of those in which the collection of safety data is ancillary to the registry's primary objectives. The legal obligations of regulated industries are discussed by others and are only mentioned briefly here. Similarly, issues to consider in the design and analysis of registries are covered in Chapters 3 and 13, respectively. Chapter 12 discusses practical and operational issues with reporting AE data from registries. The potential ethical obligations, technical limitations, and resource constraints that face registries with multiple different purposes in considering their role in AE detection and reporting are also discussed in this chapter. Case Examples 44 and 45 offer descriptions of how some registries have provided data for product safety assessments.
2. Registries Specifically Designed for Safety Assessment
Disease and product registries that systematically collect data on all eligible patients are a tremendous resource for capturing important information on safety. Registries commonly enroll patients who are not just different from but more complicated than those included in clinical trials, in terms of the complexity of their underlying disease, their comorbidities, and their concomitant medications.
2.1. Design Considerations: Disease Registries Versus Product Registries
Product registries, by definition, focus on patients treated with a particular medical product. To be useful, the registry should record specific information about the products of interest, including route of administration, dose, duration of use, start and stop date, and, ideally, information about whether a generic or branded product was used (and which brand) and/or specific information about the product. Biologic medicines and devices have their own challenges, ideally requiring information about device identifiers, production lots, and batches. Disease registries include information not only on products or procedures of interest, but also on similar patients who receive other treatments, other procedures, or no treatment for the same clinical indications. By characterizing events in the broad population with conditions of interest, disease registries can make a meaningful contribution to the understanding of AE rates by providing large, systematic data collection for target populations of interest. Their generally broad enrollment criteria allow systematic capture on a diverse group of patients, and provided that they collect information about the potential events of interest, they can be used to provide a background rate of the occurrence of these events in the affected population in the absence of a particular treatment, or in association with relevant treatment modalities for comparison. The utility of this information, of course, depends on these registries' capturing relatively specific and clear information about the events of interest among “typical” patients, and the ability of readers and reviewers to gauge how well the registries cover information about the target population of interest. Generating this kind of real-world data as part of disease registries can be informative either for the design of subsequent product registries (e.g., to establish appropriate study size estimations) or for the incorporation of new treatments into the data collection as they become available, since the data can provide useful benchmarks against which to assess the importance of any signals. Some would argue that disease registries, rather than specific product registries, are more likely to be successful in systematically collecting interpretable long-term safety data, thereby allowing legitimate comparisons, to the extent possible, across types and generations of drugs, devices, and other interventions.13
Consideration should be given during the registry design phase to inclusion/exclusion criteria, appropriate comparator groups, definitions of the exposure and relevant risk window(s), and analysis planning (see Chapter 3). Registries involving products new to the market must be cognizant of selection bias, channeling bias, and unmeasured confounding by indication. Channeling bias occurs when patients prescribed the new product are not comparable to the general disease population. For example, channeling bias occurs when sicker patients receive new treatments because they are nonresponsive to existing treatments; conversely, patients who are doing well on existing treatments are unlikely to be switched to new treatments. Unmeasured confounding can also be introduced by frailty; for example, vaccine effectiveness studies can be misleading if only healthy people get vaccinated.
In some countries, cost constraints imposed by reimbursement status (whether dictated by government agencies or private insurance) mean that new therapies are restricted to narrower populations than indicated by the approved indication. For new devices or procedures, provider learning curves and experience are additional factors that must be considered in analysis planning. Since bias is inherent in observational research, the key is to recognize and control it to the extent possible. In some cases, the potential for bias may be reduced through inclusion/exclusion criteria or other design considerations (e.g., enrollment logs) (see Chapter 3). In other cases, additional data may be collected and analytic techniques used to help assess bias (see Chapter 13). Any recognized potential for bias should be discussed in any publications resulting from the registry.
In some settings, registries are used to collect specific AEs or events of interest. Once the types of AEs and/or other special events of interest have been identified the registry must be designed to collect the data efficiently Without adequate training of clinical site staff to recognize and report events of interest, the registry will be reduced to haphazard and inconsistent reporting of AEs.
Upon registry inception, clinicians or other health care professionals who may encounter patients participating in the registry should be educated about what AEs or other special events of interest should be noted, and how and within what parameters (e.g., time) they should report untoward events that may occur while they are participating in the registry. They should also be reminded about the need to follow up on events that may not obviously be of immediate interest. For example, if a clinician asked a patient how he was feeling and the patient replied that he just returned from the hospital, it would be incumbent on the clinician to obtain additional information to determine whether this hospitalization might be a reportable event, regardless of whether the patient may have recognized it as such. This is particularly important in registries designed to capture all suspected adverse reactions as opposed to specific AEs. Such an active role by participants as well as their treating clinicians can contribute to a robust safety database. In addition to identifying events known to be of interest, the systematic collection of followup data can also capture information regarding risks not previously identified risks associated with particular subgroups (e.g., pediatric or geriatric patients, patients with liver impairment, fast or slow metabolizers), or differences in event severity or frequency not appreciated during clinical development.
Consideration should also be given to implementation of routine followup of all registry patients for key AEs, as well as vital status and patient contact and enrollment information at prespecified visits or intervals, to ensure that analyses of the occurrence of AEs among the registry population are not hampered by extensive missing data. Otherwise, the possibility that patients “lost to followup” may differ from those with repeat visits, with regard to risk of AEs, cannot be excluded.
It is also important to keep in mind that it may be necessary to revisit the registry design if it becomes apparent that the initial plan will not meet expectations. For example, the original criteria for defining the target population (patients and/or health care providers) may not yield enough patients, such as when a treatment of interest is only slowly coming into use for the intended population.
2.1.1. Health-Care Provider– and Patient-Reported Outcomes
Registries and other prospective data collection approaches have the advantage of incorporating both health care provider– and patient-reported data. Although patients and their advocates may spontaneously report postmarketing AEs to manufacturers (e.g., via inquiries directed to medical information departments) and directly to regulatory bodies, this is relatively uncommon. Furthermore, spontaneous reports received directly from patients that lack health care provider confirmation may fall outside of standard aggregating processes by regulatory bodies. In Europe, there are schemes in some countries to encourage patients to report directly to regulatory authorities; throughout Europe, manufacturers have an obligation to follow up patient reports with their health care provider. However, significant events that are not clinically recognized may be substantially underreported.
In addition, registries may collect health care provider–level data, such as training level, number of patients seen annually, and practice type and locations, that may contribute to an understanding of differences in event rates and reporting. This, along with the patient-reported data not routinely or consistently captured in the medical record (such as concomitant environmental and lifestyle exposures and adherence to prescribed regimes), differentiates registries from other electronic data sources, and in many cases allows for improved assessment of confounding and ability to assess the potential of a signal internally, prior to further signal evaluation or action.
2.1.2. Effects Observed in a Larger Population Over Time
Registries, including those used to follow former clinical trial participants, are well suited to the identification of effects that can only be observed in a large and diverse population over an extended period of time. They make it possible to follow patients longitudinally, and thereby identify long-term device failures or consequences or delayed drug safety issues or benefits; for example, failures of orthopedic implants increasingly placed in more active, younger patients. Similarly, such long-term followup facilitates evaluation of drug-drug interactions (including interactions with new drugs as they come to market and are used) and differences in drug metabolism related to genetic and other patient characteristics.
One of the most consistent risk factors for AEs is the total number of medications taken by a patient.14 Polypharmacy is commonplace, especially in the elderly, and health care providers are often unaware of over-the-counter, herbal, and other complementary (alternative) medications their patients take. Registries that collect data directly from patients can seek information about use of these products. In the case of registries used solely by health care practitioners, data collection forms can be designed specifically to request that patients be asked about such use.
When designing a registry for safety, the size of the registry, the enrolled population, and the duration of followup are all critical to ensure applicability of the inferences made from the data. If the background rate of the AE in the population of interest is not established and the time period for induction is not well understood it is extremely difficult to determine an exact meaningful target size or observation period for the registry, and the registry may be too small and have too brief an observation period to detect any, or enough, events of interest to provide a meaningful estimate of the true AE rate. In addition, the broad inclusion criteria typical of registries make it likely that subgroups of exposed patients may be identified and analyzed separately. Such stratified analyses may require larger sample sizes to achieve rate estimations with confidence intervals narrow enough to allow meaningful interpretation within strata.
As is also true for clinical trials, which often do not have a sufficient sample size for safety, but rather, for efficacy endpoints, describing safety outcomes from observational studies in statistical terms is not always straightforward. Postmarketing data may or may not confirm event rate estimates seen in clinical trials, and may also identify events not previously observed. During clinical development, risk of events not yet seen but possibly associated with a product class or the product's mechanism of action is often identified as part of ongoing risk assessment, and these events usually continue to be events of interest after approval. An inferential challenge arises when such an event is never observed. The “rule of three” is often cited as a means of interpreting the significance of the fact that a specific event is not being observed in a finite population (i.e., that the numerator of its rate of occurrence is zero). Using asymptotic risk estimation, the rule posits that in a large enough study (i.e., >30 patients), if no event occurs, and if the study were repeated over and over again, there can be 95-percent confidence that the event (or events) would not actually occur more often than one in n/3 people, where n is the number of people studied.15 The rule, originally described by Hanley and Lippman-Hand in 1983, is probably summarized best as a means for “estimating the worst case that is compatible with the observed data.”16 For the purposes of registries, this rule must be carefully applied, since it assumes that reporting of all events occurring in the study population is complete and that the study population is an accurate representation of the intended population. Nonetheless, this rule of thumb provides some guidance regarding registry size and interpretation of results.
2.1.3. Challenges
In planning a registry for safety, it is essential to consider how patients will be identified and recruited in order to understand which types of patients will be included, and equally, if not more importantly, what types of patients will likely not be included in the registry. For example, safety registries often seek information about all treated patients, regardless of whether the product is prescribed for an approved indication. While it is conceptually straightforward to design a registry that would include information on all product users, practical challenges include the difficulty of raising awareness about the existence of the registry, the desirability and importance of collecting information on all treated patients, and the challenge of specifying the AEs and other events of interest without causing undue concern about product safety.
Drawing attention to the registry among health care providers who use the treatments off label is especially challenging, due to competing concerns about being inclusive enough to capture all use (on-label or not) versus the need especially if the sponsor of the registry is also a manufacturer, to avoid the appearance of promoting off-label use when contacting physicians in specialties known to use the product off label. In addition, diseases targeted for off-label use may be markedly different from indicated uses and may pose different safety issues. In Europe, when there is limited knowledge about the safety of a product prior to its authorization and when a registry is part of a risk management plan, manufacturers may be required prior to launch of the product, to notify all physicians who may possibly prescribe the product about the existence of a registry (sometimes also called in this context a postauthorization safety study or PASS), including details of how to register patients.
It is more challenging to evaluate the utility of a registry when the entire population at risk has not been included; however, this situation merits careful consideration, since it is far more common than one where a registry captures every single treated patient. Registries organized for research purposes are typically voluntary by design, a situation that does not promote full inclusiveness.
Two key questions concern the target population (in terms of representativeness and the potential to generalize the results) and the size of the registry. When considering the target population, it is important to assess (1) whether the patients in the registry are representative of typical patients, and (2) what types of patients may be systematically excluded or not enrolled in the registry. For example, do patients come from a diverse array of health care settings or are they recruited only from tertiary referral hospitals? In the latter case the patients can be expected to be more complicated or have more advanced disease than other patients with a similar diagnosis. Are there competing activities in the target population, such as large registration trials or other observational studies, that may skew participation of sites or patients? Are patients in late stages of the disease or with greater disease severity more likely to participate? (See Chapters 3 and 13 for more information on representativeness.)
The ability to use registries for quantification of risk is highly dependent on understanding the relationship between the enrolled population and the target population. While it is intellectually appealing to dismiss the value of any registry that does not have complete enrollment of all treated patients or a documented approach to sampling the entire population, registries that can demonstrate that the actual population (the population enrolled) is representative of the target population through other means (e.g., by comparison to external data sources) can nevertheless be tremendously informative and may be the only feasible way that data can be collected.
Consider, for example, the National Registry for Myocardial Infarction (NRMI), one of the first cardiac care registries.17 NRMI was originally intended to obtain information about time to treatment for patients presenting with myocardial infarction to acute care hospitals. The program ultimately resulted in 70 publications (out of more than 500) that provided detailed information on both specific AEs for specific products and comparative information on safety events. Although this registry was quite large in terms of hospitals and patients, it included neither all MI patients nor all patients using the product for which it described safety information. It was nevertheless considered to be broadly representative of typical MI patients who presented for medical care.
2.1.4. Defining Exposure and Risk Windows
Patients will enter a registry at various stages in the course of their disease or its medical management. Therefore, it is essential to collect information on the timing of events in relation to the initial diagnosis and in relation to the timing of treatments. It is simplest to collect prespecified clinical data recorded on standardized forms at scheduled assessments, a practice that leads to uniformity within the analysis. However, many registry patients present themselves for data collection on a more naturalistic schedule (i.e., data are collected whenever the patient returns for followup care, whether or not the visit corresponds to a prespecified data collection schedule). The more haphazard schedule is more reflective of “real-world” settings, yet results in nonuniform data collection for all subjects.
Rather than being discarded these nonuniform data can be analyzed both by categorizing patient visits in terms of time windows of treatment duration (e.g., considering data from all visits occurring within 30 days of first treatment, then within 90 days, 180 days, etc.), and also by using time in terms of patient days/years of treatment. This type of analysis facilitates characterization of the type and rate of occurrence for various AEs in terms of their induction period and patient time at risk. When the collection of AE data is completed through an ongoing active process and is expected to be continued over the long term, periodic analysis and reporting should be structured around specified time points (e.g., annually, semiannually, or quarterly) and may align with the periodic safety update reports. The rigor of prespecified reporting schedules requires periodic assessment of safety and can support systematic identification of delayed effects.
In addition to variability in the timing of followup, consideration must be given to other recognized aspects of product use in the real world; for example, switching of therapies during followup, use of multiple products in combination or in sequence, dose effects, delayed effects, and failures of patient compliance. The current real-world practices for the treatment of many conditions, such as chronic pain and many autoimmune diseases, include either agent rotation schemes or frequent switching until a balance between effectiveness and tolerability is reached—practices that make it difficult to determine exposure-outcome relationships. Switching between biologics may lead to problems with immunogenicity because even products that are clinically the same, as in the case of the erythropoietins, will have different immunogenic potential due to differences in manufacturing processes and starting cell lines. In addition, as with many clinical studies, patient adherence to treatment—or lack thereof—during registry followup is an important potential confounder to consider. Over time, patients may take drug holidays and self-adjust dosages, and these actions should be, but are not always, captured via the data collected in the registry, especially if the interval between followup time points is long or the action is not known by the treating physician. Assessing the temporality of unanticipated events may then be hampered by the inability to fully characterize exposure.
Delayed effects may include late onset immunogenicity, the development of subclinical effects associated with chronic use that are not appreciated until years later, and effects that develop after stopping treatment, related to products with a long half-life or extended retention in the body. An example of this can be seen in the case of bisphosphonates used for bone resorption inhibition in the treatment of osteoporosis, where the product is retained in the bone for at least 10 years after stopping therapy, and there is some evidence that long-term bone turnover suppression puts patients at increased risk of osteonecrosis and nonspinal fractures.18 In addition, many biologics aimed at immunomodulation carry an increased risk of future malignancy that is not fully appreciated as do novel therapies directed at angiogenesis. Although registries are well suited to long-term followup, consideration must be given to how long is long enough to appreciate these effects.
Noncompliance can have a substantial effect on the assessment of AEs, particularly if dose or cumulative dose effects are suspected. Patient compliance may be affected by expense, complexity of dosing schedule, convenience/mode of administration, and misunderstanding of appropriate administration, and is not fully ascertained by data sources that capture prescriptions rather than actual product use. With products used to treat chronic diseases it is possible to estimate compliance via electronic health records, by first estimating when repeat prescriptions should be issued and then measuring the observed versus expected frequency. Although registries may be directly designed to track compliance through patient diaries and other methods of direct reporting, capturing compliance accurately and minimizing recall bias remain challenges.
2.1.5. Special Conditions: Pregnancy Registries
The use of specially designed registries for specific safety monitoring has a long history. For example, pregnancy registries are commonly used to monitor the outcomes of pregnancies during which the mother or father was exposed to certain medical products. The Antiretroviral Pregnancy Registry is an example of a registry that collects information on a broad class of products to determine the risk of teratogenesis.19 (See Case Examples 49 through 52.) Pregnancy registries provide in-depth information about the safety of one or more products and are particularly useful since, unless the product is used for life-threatening diseases or to treat a pregnancy-related illness, pregnant women are generally excluded from clinical investigations used for product approval. Registries and other observational studies, by virtue of being sustainable over longer periods of time and more amenable to small site-to-patient ratios than registration trials, can facilitate the active surveillance of safety in these populations. In addition, using computerized claims or billing data for pregnancy safety monitoring is hampered by the fact that patients often do not present early in pregnancy, by a lack of relevant data on other exposures (since these are often unrelated to reimbursement), and by difficulty linking maternal and infant records.
Therefore, direct prospective data collection remains the best source of meaningful safety data related to pregnancy. A challenge for pregnancy registries is to identify and recruit women early enough in pregnancy to obtain reliable information on treatments used during the first trimester, which is a critical time for organogenesis, and to obtain information about early pregnancy loss, since this information is not always volunteered by women. It is also important to obtain information on treatments and other putative exposures before the outcome of the pregnancy is known, to avoid selective recall of exposures by women experiencing bad pregnancy outcomes. For more information, see Chapter 21.
2.1.6. Special Conditions: Orphan Drugs
A product may be designated an orphan drug (or biologic, or medicine in the E.U.) if it fulfills certain conditions, which include being used for the diagnosis, prevention, or treatment of life-threatening or chronically debilitating conditions affecting a small number of patients. Often these diseases are extremely rare, and dossiers submitted for authorization purposes may have only tens of patients included in clinical trials. Obviously, the safety profile of such products is extremely limited, and followup of patients treated with the products after authorization is likely to be a requirement.
With some orphan drugs, the disease may have been usually fatal before therapy was available. Determining the safety profile of these products is especially difficult, in that the natural history of the disease when treated is not known, and trying to disentangle the effects of the product from those of the ongoing disease may be particularly problematic. In many of these diseases, the problem may be due to faulty enzymes in metabolic pathways, leading to accumulation of toxic substrates that cause the known manifestations of the disease. Treatment may involve blocking another enzyme or pathway, leading to the accumulation of different substances for which the effects may also not be known but are less immediately toxic. In this situation, with a fatal disease and a first product with proven efficacy, it would not be ethical to randomize patients in a trial versus placebo for an extended period of time, and so a registry may be the only effective means of obtaining long-term safety data. Registries in these situations may make meaningful contributions to understanding the natural history of the disease and the long-term effects of treatment, sometimes largely by virtue of the fact that most patients can be included and long-term followup obtained for orphan products. For more information, see Chapter 20.
2.1.7. Special Conditions: Controlled Distribution/Performance-Linked Access Systems
Registries in the United States may also be part of risk evaluation and mitigation strategies , such as restricted distribution systems, referred to as performance-linked access systems (PLAS), which may be used to monitor the safety of marketed products as one of the Elements to Assure Safe Use.20 One of the earliest PLAS was a blood-monitoring program for clozapine implemented in 1990 to prevent agranulocytosis; the program allowed clozapine to be dispensed only if an acceptable blood test had been submitted.21 Other examples include the STEPS program for thalidomide (System for Thalidomide Education and Prescribing Safety), implemented in 1998 to prevent fetal exposure;22 the TOUCH controlled distribution for natalizumab (Tysabri) for patients with multiple sclerosis, to detect the occurrence of progressive multifocal leukoencephalopathy (PML);23 and the iPLEDGE system implemented for isotretinoin in 2006, which tightly links the dispensing of isotretinoin for female patients of childbearing potential to documentation of a negative pregnancy test, to prescriber confirmation that contraceptive counseling has occurred and to prescriber and patient identification of contraceptive methods chosen.24
In many of these programs, access to the product is linked directly to participation in the related registry. Therefore, all patients treated with the product should be in the related registry because they cannot otherwise obtain access to it. The related registry is looking for a known AE (for example, PML) and collects data specifically related to that AE. The registry also collects information on other factors that may raise a patient's individual risk for this AE, information that helps provide important clinical context that would not otherwise be available in a systematic fashion on a large population of treated patients.25
While PLAS registries are driven by safety concerns, they are primarily focused on prescribing or dispensing controls rather than signal detection. As a result, they use very limited data collection forms to minimize burden, and this can limit their utility for certain types of analyses.
In Europe, use of registries for risk minimization activities can be more problematic, due to differences in national legislation and the enactment of the European Union data protection directive. In some countries it is possible to mandate registration of patients in relation to particular products (e.g., clozapine in the United Kingdom and Ireland), but in others other methods must be found. For these reasons, registries are more frequently used on a voluntary basis to monitor safety and capture AEs, while risk minimization is achieved by controlled distribution with compulsory distribution of educational material, prescribing algorithms, and treatment initiation forms to anyone likely to prescribe the product. Despite the fact that patient registration is voluntary, high enrollment rates can be achieved particularly when clinicians recognize that information on the safety profile of the product is limited.26 Obviously, if a product has a high potential for off-label use, patients enrolled in a registry may not be generalizable to all those treated with the product, but this can be factored into data analysis and interpretation. A voluntary registry coupled with controlled distribution may, in fact, be reasonably representative, since off-label use may be severely limited by difficulties obtaining the product.
2.1.8. Special Conditions: Medical Devices
Medical devices pose different analytic and data challenges from drugs. On the one hand, it is much more straightforward to identify when a device is implanted and explanted if those records can be obtained; however, since not all medical devices are covered by medical insurance, it can be more difficult to identify all the appropriate practitioners and locate all the records. Medical devices that can be attached and detached by the consumer, such as hearing aids, are very difficult to study in that, much like products used on an as-needed basis, special procedures are required to document their use; these procedures are costly and intrusive, and are therefore rarely used. Additionally, the lack of unique device identifiers has posed a challenge for safety surveillance of devices. Recently, the FDA proposed a new system that will establish a unique identifier for most medical devices, with the goals of supporting more accurate reporting and analyzing of medical device AE reports (see Chapter 23).
Despite these challenges, the safety of medical devices is very important due to their widespread use; of particular concern are long-term indwelling devices, for which recall in the event of a malfunctioning product is inherently complicated. For example, in the late 1970s/early 1980s, when a particular type of Björk-Shiley prosthetic heart valve was found to be defective and prone to fracture, leading to sudden cardiac death in the majority of cases, detailed studies of explanted devices, patient factors, and manufacturing procedures led to important information that was used to guide decisionmaking about which devices should be explanted.27, 28 Identification of the characteristics of valves at high risk of failure was very important due to the perioperative mortality risk from explanting a heart valve regardless of its potential to fail. This same logic applies to many other medical devices that are implanted and intended for long-term use. Some of the challenges relating to studying medical devices have to do with being able to characterize and evaluate the skill of the “operator,” or the medical professional who inserts or implants the device. These operator characteristics may be as, or more, important in terms of understanding risk than the characteristics of the medical devices themselves.29 For more information, see Chapter 23.
3. Registries Designed for Purposes Other Than Safety
Registries may be designed to fulfill any number of other purposes, including examining comparative effectiveness, studying the natural history of a disease, providing evidence in support or national coverage decisions, or documenting quality improvement efforts. Although these registries may gather data on AEs and report those data (to regulatory authorities, manufacturers or others), not all data may necessarily be reported through the registry. Thus, the registry may not record all events, which would result in an imprecise, and possibly inaccurate, estimation of the true risk in the exposed population(s). A strength of comparative effectiveness registries, however, lies in the systematic collection of data for both the product of interest and concomitant, internal controls.
As an example of the limitations of assessing safety events in registries not designed for safety, a registry may be sponsored by a payer to collect data on every person receiving a certain medication. The purpose of the registry may be to assess prescribing practices and determine which patients are most likely to receive this product. The registry may also contain useful data on events experienced by patients exposed to the product, but may not be considered a comprehensive collection of safety data, or may provide information regarding a known risk or outcome rather than generating data that could identify a previously unappreciated event. Alternatively, a registry may be designed to study the effectiveness of a new product among a population subset, such as the elderly. The registry may be powered to analyze certain outcomes, such as rehospitalizations for a condition or quality of life, but may not be specifically of sufficient sample size to reliably assess overall safety in this population.
It is more challenging to accurately and precisely detect AEs of interest when a registry has not been designed for a specific safety purpose. In this situation, the registry must collect a wide range of data from patients to try to catch any possible events, or be adapted later should safety become a primary objective. Some events may be missed because the registry did not anticipate them and did not solicit data to identify them. Also, much the same as for registries designed specifically to detect AEs, some events may be so rare that they do not occur in the population enrolled in the registry or do not occur during the registry followup period. In these circumstances, registries can be designed to provide useful data on some of the events that may occur in the exposed population. Such data should not be considered complete or reliable for determining event rates, but, when the data are combined with safety data from other sources, trends or signals may become apparent within the data set.
4. Ad Hoc Data Pooling
One way to capitalize on data that, because they were collected for another purpose, may be insufficient for meaningful standalone analysis and interpretation due to study size or lack of comparators, is to pool the data with other similar data. As with any pooling of disparate data, the use of appropriate statistical techniques and the creation of a core data set for analysis are critically important, and are highly dependent on consistency in coding of treatments and events and in case identification.
It is essential to have an understanding of how every data set that will be used in a pooled analysis was created. For example, what is recorded in administrative health insurance claims depends largely on what benefits are covered and how medications are dispensed. Noncovered items generally are not recorded. For example, mental health services are often contracted for under separate coverage (so-called “carve-outs”) and not covered under traditional health insurance coverage; thus, the mental health consultations are not likely to be included in administrative databases derived from billing claims data. Also, some injectable medications (e.g., certain antibiotics) may be administered in the physician's office and thus would not be recorded through commonly used pharmacy reporting systems that are based on filling and refilling prescriptions. The absence of information may lead to false conclusions about safety issues. Also, AE data coded using the same coding dictionary (e.g., Medical Dictionary for Regulatory Activities, or MedDRA) may still be plagued by inconsistency in the application of coding guidelines and standards. Recoding of verbatim event reports may be required if feasible, prior to analysis. Depending upon the purpose for which the data were collected, data on the treatments of interest are not always recorded or are not recorded with the specificity needed to understand risk (e.g., branded vs. generic, dosage, route of administration, batch).
Another consideration is differential followup, including the duration and vigor of followup in the registries to be pooled. Particular care is needed when combining data sets from different European countries, since differences in medical practice and reimbursement may mean that superficially similar data may actually represent different subgroups of an overall disease population. Similar caution is also advisable when combining information from disparate health systems within a single country, as some treatments of interest may be noncovered benefits in some systems and consequently not recorded in that health system's records. An alternative to pooling data is to conduct meta-analyses of various studies using appropriate statistical and epidemiologic methods.
While the types of registries described above may not be individually of sufficient sample size to detect safety issues, combining data from registries for other purposes could significantly enhance the ability to identify and analyze safety signals across broader populations. Core data sets for AEs have been suggested for electronic health records systems and as part of national surveillance mechanisms (e.g., through distributed research networks). In such a network, each participating registry or data source collects a standardized core data set from which results can be aggregated to address specific surveillance questions. For example, there is significant national interest in understanding the long-term outcomes of orthopedic joint implants. Currently, there are several prominent registries in the United States with varying numbers of types of patients and types of implants. Many of these registries collect data for quality improvement purposes, but have sufficient data elements to potentially report on AEs. However, only by aggregating common data sets across many of these registries can a broadly representative population be evaluated and enough data accrued to understand the safety profile of specific types of devices in particular populations.
As described above, while not every registry is designed to evaluate safety, even registries designed for other purposes might contribute to aggregate information about potential harm from health care products or services. Yet many registries, especially disease registries, are conducted by nonregulated entities such as provider associations, academic institutions, and nonprofit research groups, whose role in AE reporting is unclear. Furthermore, sample sizes needed to understand safety signals are generally much larger than those needed to achieve useful information on quality of care or the natural history of certain diseases, and the safety analyses can require a high degree of statistical sophistication. Enrolling additional patients or committing additional resources for specialized analyses in order to achieve a general societal benefit through safety reporting is not feasible for most registries when the primary purpose is not safety. However, encouraging registries to participate in aggregation of data when such participation is at minimal cost and enhances the common good may be both reasonable and appropriate.
Many efforts are underway to improve the feasibility of broader safety reporting from both registries and electronic health records that serve other purposes. These efforts include recommending standardized core data sets for safety to enhance the aggregation of information in distributed networks, and making registries interoperable with facilitated safety reporting mechanisms or other registries designed for safety.30 As facilitated reporting methodologies become more common and easier for registries to implement, there will be fewer reasons for nonparticipation. In addition, linkage of population-based registries, such as the Surveillance, Epidemiology and End Results (SEER) cancer registry program, with other data sources, such as Medicare, have proven invaluable for evaluating safety and other outcomes.
5. Signal Detection in Registries and Observational Studies
Although subject to debate, according to the World Health Organization definition, a safety signal is defined as “reported information on a possible causal relationship between an AE and a drug, the relationship being unknown or incompletely documented previously”31 Hauben and Aronson (2009) define a signal as “information that arises from one or multiple sources (including observations and experiments), which suggests a new potentially causal association, or a new aspect of a known association, between an intervention and an event or set of related events, either adverse or beneficial, which would command regulatory, societal or clinical attention, and is judged to be of sufficient likelihood to justify verificatory and when necessary, remedial actions.”32 The authors further posit that signals, following assessment, could subsequently be categorized as indeterminate, verified or refuted.
Additional attempts at defining or describing a safety signal for purposes of guiding product sponsors, regulators, and other researchers have come from various sources, including the Council for International Organizations of Medical Sciences (CIOMS), the FDA, and the United Kingdom's Medical and Healthcare products Regulatory Agency. Nelson and colleagues recently provided FDA with a comprehensive evaluation of signal detection methods for use in postmarketing surveillance, and included a discussion of “conventional Phase IV observational safety studies,” which would encompass registries, as part of a multipronged approach to surveillance.33 They noted that despite a focus on automated health care data sources, such as large health care claims databases, for primary surveillance and as the basis for FDA's Sentinel Network, the need for more detailed data regarding exposure and outcome measurement, as well as collection of relevant confounder data, will require that prospective observation studies be conducted to address prespecified safety-related hypotheses.
Establishing a threshold of effect size and robustness of data that would justify action, such as initiation of additional studies, FDA action, or changes in payer coverage, remains an important question and is unlikely to be uniformly applicable to all products and situations. A report was issued in 2010 from the CIOMS Working Group VIII, whose main goal is to harmonize the development, application, and interpretation of signal detection methods for use with drugs, vaccines, and biologics and to provide practical advice.34
Once a signal that warrants further evaluation is identified it is typically assessed based on the strength of the association between exposure and the event; biological plausibility; any evidence provided by dechallenge and rechallenge; the existence of experimental or animal models; and the nature, consistency, and quality of the data source.35 Signals may present themselves as idiosyncratic events affecting a subset of the exposed population who are somehow susceptible, events related to the pharmacological action of the drug, or increased frequency of events normally occurring in the population (such as in the example of cardiovascular events and rofecoxib). Signals may involve the identification of novel risks, or new (or more refined) information regarding previously identified risks. If an event does appear to be product related further inquiry is required to examine whether the occurrence appears to be related to a specific treatment, a combination or sequence of treatments, or a particular dosage and/or duration of use. Events with long induction periods are particularly challenging for the ascription of a causal relationship, since there are likely to be many intervening factors, or confounders, that could account for the apparent signal.
The constant challenge is to separate a potential safety signal from the “noise,” or, in other words, to detect meaningful trends and to have a basis for evaluating whether the signal is something common to people who have the underlying condition for which treatment is being administered or whether it appears to be causally related to use of a particular product. All methods currently used for signal detection have their limitations. Attempts to use quantitative, and in some cases, automated signal detection methods as part of pharmacovigilance, including data mining using Bayesian algorithms or other disproportionality analyses, are hampered by confounders and other biases inherent to spontaneously reported data.36, 37 Other methodologies also attempt to identify trends over time and include potential patterns associated with other patient characteristics, such as concomitant drug exposures.
These methods of automated signal detection lack clinical context and only draw attention to deviations from independence between product exposure and events. No conclusions regarding causality can be drawn without a further qualitative and quantitative assessment of extrinsic factors (e.g., an artificial spike in reporting due to media attention) and potential confounders; in some cases, even with quantitative and qualitative assessments, the data may be insufficient to establish causality. Depending on the original data source, it may be impossible to address these issues within the database itself and either abstracted medical record data or prospective data collection may be required to gather reliable data. The long-term followup and longitudinal data generated by many registries merits particular methodological considerations, including how often to perform testing, what threshold is meaningful for a given event, and whether that threshold changes over time.
While some registries can serve as sources of initial safety signaling or hypothesis generation, they may also be used for further investigation of a signal generated from surveillance and quantitative analysis. As an example, existing data from the Swedish Coronary Angiography and Angioplasty Registry (SCAAR), sponsored by the Swedish Health Authorities, was used to look at long-term outcomes related to bare-metal and drug-eluting stents, once it became clear through FDA-designed and other registries in the postmarket setting that off-label use was very common and that the risk of restenosis and other long-term outcomes in the real-world patient population was not fully understood. Due to the existence of comprehensive national population registries in Sweden, researchers were able to reliably combine SCAAR data, which captured unselected, consecutive angiography and percutaneous coronary intervention procedure data, with vital status and hospitalization data, to examine fatality rates and cardiac events on a population level.38 This use of procedure and national registries provides an example of how a registry that included a well-defined population allowed for safety assessments coincident with comparative effectiveness.
6. Potential Obligations for Registry Developers in Reporting Safety Issues
In considering what actual and potential obligations there are, or may be, for registries in product safety assessment, it is useful to separate the issues into several parts. First, there are two key questions that can be asked for each registry: (1) What is the role of registries not designed for safety purposes with respect to the search for AEs? and (2) What are the obligations, especially for those registries not sponsored by regulated manufacturers, to further investigate and report these events when found? As discussed above, registries can be classified by whether or not they were designed for a safety purpose, and also by whether or not they have specified regulatory obligations for reporting. Beyond these distinctions, several factors need to be considered including the ethical obligations of the registry developer, the technical limitations of the signal detection, and resource constraints.
Registries designed for safety assessment purposes should have a clear and deliberate plan in place, not only for detecting the signal of interest, but for handling unanticipated events and reporting them to appropriate authorities. Only in the case of registries supported by the regulated industries are rules for reporting drug or device AEs explicit. Therefore, it would be helpful if other registries would also formulate plans that ensure that appropriate information will reach the right stakeholders, either through reporting to the manufacturer or directly to the regulator, in a timely manner similar to those required by the regulated industries. There should not be two different standards for reporting information intended to safeguard the health and well-being of all.
Registries that are not designed specifically for safety assessment purposes, particularly those that are not sponsored by a manufacturer, raise more complex issues. While researchers have an obligation to the patients enrolled in any research activity to alert them should information regarding potential safety issues become known, it is less clear how far this obligation extends. In the United Kingdom, the General Medical Council includes in its advice on “Good Medical Practice” the requirement to “report suspected adverse drug reactions in accordance with the relevant reporting scheme.”39 It is therefore clear that in the United Kingdom contributing to the safety profile of a medicine is regarded as part of the duties of a medical practitioner. During its review of research registries, an institutional review board (IRB) in the United States or ethics committee (EC) in Canada or the European Union may specify the creation of an explicit incidental findings plan prior to approval. Such a plan is often part of studies producing or compiling nonclinical imaging and genetic data. In addition, some investigators will have an obligation to report to an IRB or EC any unanticipated problems involving risks to subjects or others under the regulations on human research protections. In turn, IRBs and ECs have an obligation to report such incidents to relevant authorities.
At a minimum, all registries should ensure that standard reporting mechanisms for AE information are described in the registry's procedural documents. These mechanisms should also be explained to investigators and, where feasible, their reporting efforts should be facilitated. For example, all registries in the United States can make available to registry participants access to the MedWatch forms40 and train them in the appropriate use of these forms to report spontaneous events. As described in Section 4 above, in the near future it should be possible for registries that collect data electronically to actually facilitate the reporting of AEs by linking with facilitated safety reporting mechanisms. This mechanism is attractive because it reduces the work of the investigator in generating the report and ensures that the report will go to a surveillance program prepared to investigate and manage both events and potential safety signals.
Obligations beyond facilitation are less clear. Furthermore, there are both technical and resource obstacles to thoroughly investigating potential signals, and risks that inaccurate and potentially injurious information will be generated. For example, publicizing product safety issues can result in some patients discontinuing use of potentially life-saving products regardless of the strength of the scientific evidence. As described earlier, registries designed for safety assessment should ideally have both adequate sample size and signal evaluation expertise in order to assess safety issues. Registries not designed for safety purposes may not have enough patients or statistical signal detection expertise to investigate potential signals, or may not have the financial resources to devote to unplanned analyses and investigations. It would seem that, at a minimum, registries not designed for safety purposes should use facilitated reporting (via training, providing forms, etc.) of individual events through standard channels to meet their ethical obligations, and that they should check with any institutions with which they are affiliated to determine whether they are subject to additional reporting requirements. However, should a registry identify potential signals through its own analyses, obligations arise.
While registries that are approved by IRBs report safety issues to those IRBs, incidental analytic findings, which may represent true or false signals, may need more definition and should best be further investigated and reported for the public good. One approach would be to report summary information to the relevant regulatory authority for further evaluation. To avoid doubt, registry developers should consider these issues carefully during the planning phase of a registry, and should explicitly define their practices and procedures for AE detection and reporting, their planned analyses of AEs, and how incidental analytic findings will be managed. Such a plan should lay out the extent to which registry owners will analyze their data for AEs, the timing of such analyses, what types of unanticipated issues will be investigated internally, what thresholds would merit action, and when information will be provided to regulators or other defined government entities, depending on the nature of the safety issue.
7. Summary
The ongoing challenge, in the use both of existing data and of prospective data collection efforts such as registries, is to cast a wide enough net to capture not only rare events, but also more common events and events that are not anticipated (i.e., not part of a preapproval or postapproval potential risk assessment). In some cases, existing registries may add additional data collection to address questions regarding possible AEs that arise after registry initiation. In addition, it must be considered that all observational data sources are only as strong as their ability to measure and control for potential biases, including confounding and misclassification.
Large registries, linkage and distributed network schemes, and sentinel surveillance are all tools being actively developed to create an integrated approach to medical product safety and specifically, to signal detection and verification.
In contributing to the evidence hierarchy surrounding the generation of signals for detection and confirmation of potential AEs, registries are likely to make their strongest contributions through: detection of novel AEs associated with product use as reported by treating physicians, which, in turn, constitutes a signal necessitating further study; gathering information about pregnant women and other hard-to-study subpopulations of product users; linking with additional data sources such as the Medicare-SEER data linkage, thereby broadening the range of questions that can be addressed beyond the constraints of data collected for a registry; and confirming or validating signals generated in other data, such as from automated signal generation in large claims databases. Ideally, a clear and prospective understanding among stakeholders is needed regarding whether and under what circumstances signal monitoring within registries is appropriate; the timing or periodicity of any such analyses; what should be done with the information once it is identified, and what, if any, are the ethical obligations to collect, analyze, and report safety information if doing so is not a planned objective of the registry, and if the registry sponsor is not directly required by regulation to conduct such reporting.
Thoughtfully designed registries can play important roles in these newly emerging strategies to use multiple available data sources to generate and strengthen hypotheses in product safety.
However, as with all data sources, it is important to assess the effects of registry design, the type of data, reason for the data collection, how the data were collected, and the generalizability to the target population, in order to assess the strengths, weaknesses, and validity of the results provided and their contribution to the knowledge of the safety profile of the medicine or device under study.
Case Examples for Chapter 19
Case Example 44Using a registry to assess long-term product safety
Description | The British Society for Rheumatology Biologics Register is a prospective observational study conducted to monitor the routine clinical use and long-term safety of biologics in patients with severe rheumatoid arthritis and other rheumatic conditions. The United Kingdom-wide national project was launched after the introduction of the first tumor necrosis factors (TNF) alpha inhibitors. |
---|---|
Sponsor | The British Society for Rheumatology (BSR) commissioned the registry, which receives restricted funding from Abbott Laboratories, Biovitrum, Schering Plough, Roche, and Wyeth Pharmaceuticals. The registry is managed by the BSR and the University of Manchester. |
Year Started | 2001 |
Year Ended | Ongoing |
No. of Sites | All consultant rheumatologists in the United Kingdom who have prescribed anti-TNF therapy participate. |
No. of Patients | More than 17,000 |
Challenge
Rheumatoid arthritis (RA) is a progressive inflammatory disease characterized by joint damage, pain, and disability. Among the pharmacologic treatments, nonbiologic disease-modifying antirheumatic drugs (DMARDs) are considered the first-line treatment. Novel biologic therapies represent a new class of agents that prevent inflammation and have demonstrated efficacy in RA patients. The most commonly used biologics are tumor necrosis factors (TNF) inhibitors (etanercept, infliximab, and adalimumab). However, results from clinical trials and pharmacovigilance studies have raised potential safety concerns, and limited long-term data on these therapies are available. Of particular concern has been an increase of tuberculosis observed in patients treated with anti-TNF therapy.
Proposed Solution
A prospective observational registry was launched in 2001 to monitor the safety of new biologic treatments. The registry collects data on response to treatment and potential AEs every six months, and patients are followed for the life of the registry. Over 4,000 patients are enrolled for each of the anti-TNF agents (etanercept, infliximab, and adalimumab), and the registry represents approximately 80 percent of RA patients treated with these biologics in the United Kingdom. In addition to patients receiving anti-TNF therapy, the registry has enrolled a control cohort of patients receiving nonbiologic DMARDs.
Results
Data from the registry were analyzed to determine whether an increased risk of tuberculosis existed in RA patients treated with anti-TNF therapy (Dixon et al., 2010). In more than 13,000 RA patients included up to April 2008, 40 cases of tuberculosis were observed in the anti-TNF cohort and no cases in the DMARD group. A differential risk was reported among the three anti-TNF agents, with the lowest risk observed in the etanercept group. The incidence rates were 144, 136, and 39 cases per 100,000 person-years for adalimumab, infliximab, and etanercept, respectively. In addition, the incidence rate ratio, median time to events, and influence of ethnicity were evaluated.
Key Point
As novel drugs and treatments are developed and licensed registries may be useful tools for collecting long-term data to assess known and emerging safety concerns.
For More Information
Dixon WG, Hyrich KL, Watson KD. Drug-specific risk of tuberculosis in patients with rheumatoid arthritis treated with anti-TNF therapy: Results from the British Society for Rheumatology Biologics Register (BSRBR). Annal Rheum Dis. 2010;69(3):522–8 [PMC free article: PMC2927681] [PubMed: 19854715].
Zink A, Askling J, Dixon WG, et al. European biological registers: methodology, selected results and perspectives. Annal Rheum Dis. 2009;68:1240–6 [PubMed: 18647854].
Case Example 45Using a registry to monitor long-term product safety
Description | SINCERE™ (Safety in Idiopathic arthritis: NSAIDs and Celebrex Evaluation Registry) was a multi-center registry designed to monitor the long-term safety of nonsteroidal anti-inflammatory drugs (NSAIDs) in patients with juvenile idiopathic arthritis (JIA). The registry included patients ages 2 to 17 and collected demographic, developmental, clinical, and safety data. The followup period was at least 2 years, and as long as 4 years for some patients. |
---|---|
Sponsor | Pfizer, Inc. |
Year Started | 2009 |
Year Ended | 2012 (terminated early) |
No. of Sites | 16 sites in the United States |
No. of Patients | Planned enrollment of 200 patients on celecoxib and 200 patients on other NSAIDs. Actual enrollment of 219 patients on other NSAIDs, 55 on celecoxib, for a total of 274. |
Challenge
Nonsteroidal anti-inflammatory drugs (NSAIDs) have been used for more than 30 years to relieve pain and inflammation in juvenile idiopathic arthritis (JIA), and it is estimated that 80 to 90 percent of JIA patients will use an NSAID at some point. However, little is known about the long-term safety of chronic use of NSAIDs in children with JIA. This question is particularly important, as many children with JIA will continue to use NSAIDs well into adulthood. Due to the rarity of JIA and the special ethical issues surrounding children's participation in experimental studies, randomized controlled trials of NSAIDs in JIA are considerably smaller and of shorter duration than adult arthritis trials; the pivotal trial for celecoxib in JIA, one of the largest NSAID JIA studies, had 100 patient-years of exposure. In addition, randomized trials may not be generalizable to typical JIA populations. Lastly, it is unclear if the emerging safety concerns in adult NSAID and celecoxib users translate to children, who are much less likely to develop serious cardiovascular thromboembolic events or gastrointestinal bleeding events.
The development of a long-term observational study was necessary to address these knowledge gaps, fulfill a postmarketing safety commitment, and respond to concerns of regulators, patients, physicians, and the sponsor.
Proposed Solution
This multicenter registry was designed to gather long-term safety data on NSAIDs use in children with JIA, and was intended to enroll a quasi-inception cohort of patients aged 2 to 17 years and >10 kg who were prescribed (not more than 6 months prior) either celecoxib (n=200) or other NSAIDs (n=200). Pediatric rheumatologists from 16 sites in the United States entered data quarterly for the first 12 months and twice annually thereafter. The registry intended to follow all patients for at least 2 years and as long as 4 years, as all patients were encouraged to remain in the registry until the last patient completed the minimum followup. Concomitant medications and treatment switches were permitted and patients were followed for residual effects even if NSAID treatment was discontinued.
Targeted events of interest (i.e., cardiovascular, gastrointestinal, and hypertension) and general safety serious and nonserious AEs were collected in a systematic manner. The Common Terminology Criteria for Adverse Events (CTCAE ver 3.0) criteria were used to both code and grade all AEs to minimize variability across physicians. In designing the registry, particular attention was paid to collecting potential covariates relevant to confounding by indication, given the expected differential prescribing between celecoxib and other NSAIDs. The analyses summarized the incidence of the targeted events and AEs in general, and exploratory analyses may further characterize AE rates by other clinical and demographic factors.
Results
The registry was terminated early due to low patient recruitment, despite multiple attempts to improve site and patient enrollment. The primary issue identified as a barrier to enrollment was that the treatment paradigm had changed since celecoxib was first approved: with the advent and increasing use of biologic therapies, NSAIDs were no longer being used long-term in JIA very often. As a result, the objective of the study, to assess the long-term safety of celecoxib as used for JIA, could not be met. This change in treatment paradigm, coupled with safety information from the registry and other sources that indicated no new signals nor change to the benefit-risk profile of celecoxib as used in children with JIA, allowed the FDA to release the sponsor from the commitment and terminate the study early due to futility. The registry nevertheless provided over 410 patient-years of observation in this cohort of NSAID and celecoxib users, providing additional safety data on these drugs as used for JIA in routine clinical practice; no new safety issues were identified. This information may facilitate appropriate therapeutic decisionmaking for doctors and patients.
Key Point
Registries may be useful tools for examining long-term product safety, particularly in populations such as children that are difficult to study in randomized controlled trials. Changes in the treatment paradigm of a disease may affect the utility and feasibility of a long-term product safety registry.
For More Information
Beukelman T, Patkar NM, Saag KG, et al. 2011 American College of Rheumatology recommendations for the treatment of juvenile idiopathic arthritis: initiation and safety monitoring of therapeutic agents for the treatment of arthritis and systemic features. Arthritis Care Res. 2011 Apr;63(4):465–82 [PMC free article: PMC3222233] [PubMed: 21452260].
References for Chapter 19
- 1.
- Lecutier MA. Phocomelia and internal defects due to thalidomide. Br Med J. 1962 Dec 1;2(5317):1447–8. [PMC free article: PMC1926787] [PubMed: 13929004]
- 2.
- Herbst AL, Ulfelder H, Poskanzer DC. Adenocarcinoma of the vagina. Association of maternal stilbestrol therapy with tumor appearance in young women. N Engl J Med. 1971 Apr 15;284(15):878–81. [PubMed: 5549830]
- 3.
- Hartzema AG, Tilson HH, Chan KA, editors. Pharmacoepidemiology and Therapeutic Risk Management. Cincinnati: Harvey Whitney Books; 2008.
- 4.
- Drazen JM, Rainey J, Begg H, et al. Adverse Drug Event Reporting: The Roles of Consumers and Health-Care Professionals: Workshop Summary. Washington, DC: National Academies Press; 2007.
- 5.
- McClellan M. Drug safety reform at the FDA— pendulum swing or systematic improvement? N Engl J Med. 2007 Apr 26;356(17):1700–2. [PubMed: 17435081]
- 6.
- Eland IA, Belton KJ, van Grootheest AC, et al. Attitudinal survey of voluntary reporting of adverse drug reactions. Br J Clin Pharmacol. 1999 Oct;48(4):623–7. [PMC free article: PMC2014371] [PubMed: 10583035]
- 7.
- Moore N, Hall G, Sturkenboom M, et al. Biases affecting the proportional reporting ratio (PPR) in spontaneous reports pharmacovigilance databases: the example of sertindole. Pharmacoepidemiol Drug Saf. 2003 Jun;12(4):271–81. [PubMed: 12812006]
- 8.
- Figueiras A, Herdeiro MT, Polonia J, et al. An educational intervention to improve physician reporting of adverse drug reactions: a cluster-randomized controlled trial. JAMA. 2006 Sep 6;296(9):1086–93. [PubMed: 16954488]
- 9.
- European Medicines Agency. Guideline on the use of statistical signal detection methods in the Eudravigilance Data Analysis System. 2008. [September 30, 2013]. http:
//eudravigilance .emea.europa.eu/human/docs/10646406en .pdf. - 10.
- Finney DJ. The Design and Logic of a Monitor of Drug Use. J Chronic Dis. 1965 Jan;18:77–98. [PubMed: 14252273]
- 11.
- U.S. Food and Drug Administration. The Sentinel Initiative: national strategy for monitoring medical product safety. May, 2008. [August 17, 2012]. www
.fda.gov/downloads /Safety/FDAsSentinelInitiative /UCM124701.pdf. - 12.
- Samuel FE Jr. Safe Medical Devices Act of 1990. Health Aff (Millwood). 1991 Spring;10(1):192–5. [PubMed: 2045049]
- 13.
- Schmitt-Egenolf M. Psoriasis therapy in real life: the need for registries. Dermatology. 2006;213(4):327–30. [PubMed: 17135739]
- 14.
- Tulner LR, Frankfort SV, Gijsen GJ, et al. Drug-drug interactions in a geriatric outpatient cohort: prevalence and relevance. Drugs Aging. 2008;25(4):343–55. [PubMed: 18361544]
- 15.
- Hanley JA, Lippman-Hand A. If nothing goes wrong, is everything all right? Interpreting zero numerators. JAMA. 1983 Apr 1;249(13):1743–5. [PubMed: 6827763]
- 16.
- Eypasch E, Lefering R, Kum CK, et al. Probability of adverse events that have not yet occurred: a statistical reminder. BMJ. 1995 Sep 2;311(7005):619–20. [PMC free article: PMC2550668] [PubMed: 7663258]
- 17.
- Rogers WJ, Bowlby LJ, Chandra NC, et al. Treatment of myocardial infarction in the United States (1990 to 1993). Observations from the National Registry of Myocardial Infarction. Circulation. 1994 Oct;90(4):2103–14. [PubMed: 7923698]
- 18.
- Odvina CV, Zerwekh JE, Rao DS, et al. Severely suppressed bone turnover: a potential complication of alendronate therapy. J Clin Endocrinol Metab. 2005 Mar;90(3):1294–301. [PubMed: 15598694]
- 19.
- Tilson HH, Doi PA, Covington DL, et al. The antiretrovirals in pregnancy registry: a fifteenth anniversary celebration. Obstet Gynecol Surv. 2007 Feb;62(2):137–48. [PubMed: 17229330]
- 20.
- Food and Drug Administration Amendments Act of 2007, Pub. L. No. 110-85 (2007), Title IX.
- 21.
- Peck CC. FDA's position on the clozaril patient management system. Hosp Community Psychiatry. 1990;41(8):876–7. [PubMed: 2401475]
- 22.
- Zeldis JB, Williams BA, Thomas SD, et al. S.T.E.P.S.: a comprehensive program for controlling and monitoring access to thalidomide. Clin Ther. 1999 Feb;21(2):319–30. [PubMed: 10211535]
- 23.
- Biogen Idec. The TYSABRI TOUCH® Prescribing Program. [September 30, 2013]. http://www
.tysabri.com /safety-with-tysabri.xml. - 24.
- Honein MA, Lindstrom JA, Kweder SL. Can we ensure the safe use of known human teratogens?: The iPLEDGE test case. Drug Saf. 2007;30(1):5–15. [PubMed: 17194167]
- 25.
- Kleinschmidt-DeMasters BK, Tyler KL. Progressive multifocal leukoencephalopathy complicating treatment with natalizumab and interferon beta-1a for multiple sclerosis. N Engl J Med. 2005 Jul 28;353(4):369–74. [PubMed: 15947079]
- 26.
- Humbert M, Segal ES, Kiely DG, et al. Results of European post-marketing surveillance of bosentan in pulmonary hypertension. Eur Respir J. 2007 Aug;30(2):338–44. [PubMed: 17504794]
- 27.
- Walker AM, Funch DP, Sulsky SI, et al. Patient factors associated with strut fracture in Bjork-Shiley 60 degrees convexo-concave heart valves. Circulation. 1995 Dec 1;92(11):3235–9. [PubMed: 7586309]
- 28.
- Walker AM, Funch DP, Sulsky SI, et al. Manufacturing characteristics associated with strut fracture in Bjork-Shiley 60 degrees Convexo-Concave heart valves. J Heart Valve Dis. 1995 Nov;4(6):640–8. [PubMed: 8611980]
- 29.
- Curtis JP, Luebbert JJ, Wang Y, et al. Association of physician certification and outcomes among patients receiving an implantable cardioverter-defibrillator. JAMA. 2009 Apr 22;301(16):1661–70. [PMC free article: PMC2805129] [PubMed: 19383957]
- 30.
- The * ASTER Pilot Project: Improving the Reporting of Adverse Events. *ASTER: A Collaborative Study to Improve Drug Safety. [August 15, 2012]. www
.asterstudy.com. - 31.
- World Health Organization's International Drug Monitoring Programme. Uppsala Monitoring Centre (UMC). [August 15, 2012]. http://www
.who-umc.org/ - 32.
- Hauben M, Aronson JK. Defining ‘signal’ and its subtypes in pharmacovigilance based on a systematic review of previous definitions. Drug Saf. 2009;32(2):99–110. [PubMed: 19236117]
- 33.
- Nelson J, Cook A, Yu O. Evaluation of signal detection methods for us in prospective post-licensure medical product safety surveillance. Mar, 2009. [August 15, 2012]. http://www
.fda.gov/OHRMS /DOCKETS/98fr/FDA-2009-N-0192-rpt .pdf. - 34.
- Council for Interntational Organizations of Medical Sciences (CIOMS). Practical aspects of signal detection in pharmacovigilance. Report of CIOMS Working Groups VIII. Geneva: 2010.
- 35.
- Meyboom RH, Egberts AC, Edwards IR, et al. Principles of signal detection in pharmacovigilance. Drug Saf. 1997 Jun;16(6):355–65. [PubMed: 9241490]
- 36.
- Hauben M, Zhou X. Quantitative methods in pharmacovigilance: focus on signal detection. Drug Saf. 2003;26(3):159–86. [PubMed: 12580646]
- 37.
- Szarfman A, Machado SG, O'Neill RT. Use of screening algorithms and computer systems to efficiently signal higher-than-expected combinations of drugs and events in the US FDA's spontaneous reports database. Drug Saf. 2002;25(6):381–92. [PubMed: 12071774]
- 38.
- Lagerqvist B, James SK, Stenestrand U, et al. Long-term outcomes with drug-eluting stents versus bare-metal stents in Sweden. N Engl J Med. 2007 Mar 8;356(10):1009–19. [PubMed: 17296822]
- 39.
- General Medical Council. Good Medical Practice. 2006. [August 15, 2012]. http://www
.gmc-uk.org /guidance/good_medical_practice.asp. - 40.
- U.S. Food and Drug Administration. Reporting Serious Problems to the FDA. [August 15, 2012]. http://www
.fda.gov/Safety /MedWatch/HowToReport/default.htm.
- Introduction
- Registries Specifically Designed for Safety Assessment
- Registries Designed for Purposes Other Than Safety
- Ad Hoc Data Pooling
- Signal Detection in Registries and Observational Studies
- Potential Obligations for Registry Developers in Reporting Safety Issues
- Summary
- Case Examples for Chapter 19
- References for Chapter 19
- Use of Registries in Product Safety Assessment - Registries for Evaluating Patie...Use of Registries in Product Safety Assessment - Registries for Evaluating Patient Outcomes
- RecName: Full=Cytochrome b-c1 complex subunit 6, mitochondrial; AltName: Full=Co...RecName: Full=Cytochrome b-c1 complex subunit 6, mitochondrial; AltName: Full=Complex III subunit 6; AltName: Full=Complex III subunit VIII; AltName: Full=Cytochrome c1 non-heme 11 kDa protein; AltName: Full=Mitochondrial hinge protein; AltName: Full=Ubiquinol-cytochrome c reductase complex 11 kDa protein; Flags: Precursorgi|20455515|sp|P07919.2|QCR6_HUMANProtein
Your browsing activity is empty.
Activity recording is turned off.
See more...