Drug Safety Priorities: Initiatives and Innovation
Drug Safety Priorities: Initiatives and Innovation
Drug Safety Priorities: Initiatives and Innovation
www. fda.gov
CONTENTS
This report goes beyond previously issued safety-related reports to offer a broader picture
of FDA’s safety efforts, highlighting the critical advantages gained through a range of
multidisciplinary collaborations and partnerships. From innovative computing platforms that
support new drug reviews, to the ongoing implementation of existing programs like Safety
First and the Safe Use Initiative, the active surveillance capacities of the Sentinel System,
improvements in drug product quality oversight, and state-of-the-art communications tools that
exploit new opportunities in social media and mobile device applications, FDA is evolving
a drug safety enterprise that leverages gains achieved within the Agency as well as through
alliances with our federal partners and with numerous stakeholder organizations.
If the resulting evidence from early development (laboratory tests and animal
studies), and all human clinical trials support a drug’s safety and efficacy for its
intended use, a company can file a New Drug Application (NDA) to market the
drug.
DRUG DEVELOPED
Drug sponsor develops a new drug compound and seeks to have it approved by FDA for sale in the United States.
IND APPLICATION
The sponsor submits an Investigational New Drug (IND) application to FDA. The
application includes information about the drug’s composition and manufacturing,
results from initial testing, and a plan for testing the drug on humans.
PHASE 1 CLINICAL
Phase 1 studies are usually conducted in healthy volunteers. The Phase 1 Drug Sponsor’s Clinical
emphasis is safety. The goal is to determine what the drug's most frequent Studies/Trials
side effects are and, often, how the drug is metabolized and excreted. The
number of subjects typically ranges from 20 to 80.
PHASE 2
Phase 2 studies begin if Phase 1 studies don't reveal unacceptable
toxicity. The Phase 2 emphasis is effectiveness. The goal is to obtain
preliminary data on whether the drug works in people as a treatment for
a certain disease or condition. Safety continues to be evaluated, and
short-term side effects are studied. The typical number of subjects in
Phase 2 studies ranges from a few dozen to about 300.
PHASE 3
Phase 3 studies begin if evidence of effectiveness is shown in Phase 2,
gathering more information about safety and effectiveness, studying the
drug in different populations and at different dosages, and in combination
with other drugs. The number of subjects in Phase 3 studies usually
ranges from several hundred to about 3,000 people.
NDA APPLICATION
The drug sponsor formally asks FDA to approve a drug for marketing in the United
FDA DRUG
SPONSOR
States by submitting an NDA. An NDA includes all animal and human data and analyses
of the data, as well as information about how the drug behaves in the body and how it
is manufactured.
APPLICATION REVIEWED
After an NDA is received, FDA has 60 days to decide whether to file it so it can be reviewed.
If FDA files the NDA, the FDA Review Team is assigned to evaluate the sponsor’s research on
the drug’s safety and effectiveness.
DAY
DRUG LABELING
FDA reviews the drug’s professional labeling and assures appropriate information is
communicated to health care professionals and consumers.
60
FACILITIES INSPECTED
FDA inspects the facilities where the drug will be manufactured.
POSTMARKETING SURVEILLANCE
Tracking of safety issues, required
postmarketing studies or clinical trials
FDA
APPROVED
DRUG APPROVAL
FDA approves the application or issues a complete response letter.
www.fda.gov
In providing the operational structure for evaluating and acting on drug safety
issues in the postmarket period, Safety First supports and reinforces safety
oversight across the product lifecycle and serves as an integral aspect of
CDER’s overall drug safety program.
Drug safety
science includes
the development of
Drug safety science, which includes the development of tools, techniques, tools, techniques,
and data sources that help CDER to better identify and manage safety issues and data sources that
related to drug effects and product quality, is an integral component of FDA’s help CDER to better
regulatory science portfolio. As profound new insights into disease processes
at cellular and molecular levels operate in the technologically advanced
identify and manage
environments of distributed digital networks, data mining, custom-built safety issues.
software platforms, and mobile device applications, drug safety science is
taking on ever more powerful potential to predict, detect, and prevent drug-
related adverse events.
As pharmaceutical developers and manufacturers submit electronic product applications in ever greater numbers,
the applications have also grown in complexity. In turn, CDER’s workload has increased, and digital tools used to
analyze large amounts of data are critical to consistent, reliable, and expeditious assessments of drug safety and
efficacy.
The scientists and physicians who serve on CDER’s review teams, although experts in their respective specialty
areas, may have difficulties and spend valuable time navigating the digital intricacies of electronic drug product
submissions. JumpStart, a vital new service in CDER’s portfolio, is meeting this challenge.
JumpStart is a software platform that provides an early assessment of data quality and product safety to
CDER’s review staff, enabling reviewers to spend less time manipulating complex data and more time
applying their expertise to identify safety and efficacy concerns and support informed decision making.
More computing tools have been incorporated into the JumpStart service—combining automated
analyses for data fitness and safety. Next steps include the piloting of additional tools and services,
expanding data fitness assessment capabilities, and building analysis “libraries” to include more
analyses aligned with specific areas of drug review.
JumpStart Updates—2015-2016
JumpStart has been offered to CDER’s drug review teams for the third year, providing reviewers
with better understanding of submission data earlier in the review process, highlighting areas of
potential concern and facilitating the efficiency of safety signal identification and risk analysis.
• Thirty full service JumpStart training sessions were provided for drug reviewers. Based on
feedback, the JumpStart team improved the data fitness sessions to better meet the needs of
reviewers.
• Work proceeded on assessing the feasibility of integrating clinical trials data with JumpStart.
The Janus Clinical Trials Repository (CTR) is a data warehouse that can provide access to study
data in ways that reviewers need, whether as an individual study or a set of studies integrated
for safety assessments. OCS also explored the use of CTR to facilitate analyses of safety risk across
classes of drugs or focused on specific treatment indications.
• A robust training program continues to ensure that newly developed digital tools are
seamlessly adopted across the review community to improve pre-market safety assessments.
Voluntary Voluntary
Regulatory Requirements
FAERS
Database FDA
FAERS helps to bridge the gap between known AEs seen in pre-approval clinical trials and those seen
once an approved drug is used in the general population. If a potential safety concern is identified in
FAERS, further evaluation is conducted on a systematic basis. Monitoring of individual spontaneous
reports and aggregate data from the FAERS database for all marketed products is conducted weekly.
Reviews of AE reports and benefit-risk evaluation reports occur at quarterly, bi-annual and annual
intervals, and full drug product safety reviews are conducted 18 months after approval. CDER employs
data mining techniques to efficiently identify FAERS reports of greatest value, and is continuously
working to update these approaches.
When reviewing reports, particular attention is paid to those AEs that are both serious and unexpected.
An important part of the review focuses on the narrative submitted by the person reporting the AE. The
more complete a report’s medical and clinical information is, the more useful the report will be, and
reporters are encouraged to include as much detail as possible in their narratives.
When a safety concern is detected, further evaluation might include studies conducted using other large
databases such as those housed in the Sentinel System, the Centers for Medicare and Medicaid Services,
and the Veterans Health Administration, among others. Depending on the results of additional evaluation,
CDER may take regulatory action to improve product safety and protect the public health, such as
updating a product’s labeling information, called a Safety Labeling Change, communicating new safety
information to the public, or, in rare cases, removing a product from the market.
Even though CDER receives more than one million AE reports every year, not all AEs associated with
a given drug are reported. Many factors influence whether or not an event will be reported, such as the
time a product has been on the market or the nature of publicity about a drug-related event. Because
AEs are substantially underreported, FAERS data cannot be used to calculate the incidence of a specific
AE or other problem in the U.S. population. It is also understood that reports submitted to FAERS are
a highly selective sample of the events observed in real-world medical practice, which are frequently
confounded by factors such as multiple drug use and worsening of pre-existing disease. As a result, it is
rarely, if ever, certain that a reported AE was actually caused by an associated drug product. CDER does
not require that a causal relationship between a product and event be proven before reporting, and reports
may not always contain enough detail to properly evaluate the event. Despite these limitations, FAERS
remains a very useful surveillance tool that CDER is actively working to improve.
In 2016 alone, the FAERS database will have logged nearly two million AE reports, and now contains
more than 12 million reports.
Non-Expedited
1,200,000
15-Day
Direct
1,000,000
Number of Reports
800,000
600,000
400,000
200,000
0
2004 2005 2006 2007 2008 2009 2010 2011 2012 2013 2014 2015
This graph shows the number of direct, 15-day, and non-expedited (also known as periodic) reports received and
entered into FAERS from 2004 through 2015. FDA receives direct reports from the public while 15-day and non-
expedited reports are submitted by industry. The 15-day reports describe AEs from spontaneous reports that are
serious and unexpected, as well as AEs from clinical trials that are serious, unexpected and judged to be reasonably
associated with the drug. Industry submits all other adverse event reports as non-expedited reports.
An important foundation of the Sentinel System is its collaborative nature and active engagement with
a broad range of stakeholders, including academic medical centers, healthcare systems, health insurers,
and patient advocacy groups. With data analytics leveraged across broad digital networks, and with
access to information provided by data partners, including electronic health record systems (EHRs),
administrative and insurance claims databases, and registries, Sentinel enables FDA to obtain vital
data to help evaluate safety issues—and demonstrates the benefits that can be achieved through broad
collaboration.
In addition to its vital contribution to drug safety surveillance, the Sentinel System is also an invaluable
national scientific resource for a broad range of other applications. A current goal is that, working with
other scientific groups, a National Data Infrastructure can be created that enables many users (such
as other governmental agencies or researchers from academia or industry) to access various types of
digital data for various purposes, including evaluating medical product performance and healthcare
quality improvement.
• The Active Risk Identification and Analysis (ARIA) System was integrated into FDA’s day-
to-day regulatory and review activities. A subcomponent of the Sentinel System, ARIA
leverages existing analysis methods and Sentinel’s Common Data Model to evaluate safety
signals identified during both premarketing and postmarketing settings.
• CDER used Sentinel’s rapid query capability in ARIA to run nearly 2,000 different medical
product scenarios, and to access the electronic healthcare data (with privacy controls in
place) of over 193 million health plan members.
• CDER continued to coordinate with the Center for Biologics Evaluation and Research to
expand their vaccine and blood products surveillance capabilities under their Postmarket
Rapid Immunization Safety Monitoring (PRISM) and Blood Surveillance Continuous Active
Network (BloodSCAN), and with the Center for Devices and Radiological Health to
complement the National Evaluation System for Medical Devices (NESMD).
• Blue Cross/Blue Shield of Massachusetts and the Hospital Corporation of America (HCA)
came aboard as Sentinel data partners. HCA provides access to data from inpatient
hospitalizations, including inpatient medications and transfusions, expanding Sentinel’s
reach and capabilities.
priority gaps that could be addressed through targeted research projects. These to confirm a diagnosis,
priorities are assessed within the broad framework of FDA’s overall regulatory predict a likely medical
science objectives and evolving needs which help guide development of outcome, or monitor a drug’s
CDER Fellowships
Offices
Contracts
and Grants
Mechanisms facilitating and supporting CDER safety research efforts include contributions from other Federal
agencies, public-private partnerships, academic collaborations, funding through grants and contracts, and
the ongoing expertise provided by CDER’s scientific offices and divisions.
• Insights into the links between a drug’s chemical structure and its potential to induce adverse events
• Research-based predictions that can be used to support decisions for new and generic drugs when
safety data are limited
DARS conducts studies that cannot or will not be done by industry or other Federal agencies, with a
staff that provides technical capabilities using a range of tools to predict such safety-related outcomes
as genotoxicity, carcinogenicity, cardiotoxicity and drug-induced liver injury. DARS consultations assist
CDER safety reviewers in the interpretation of data submitted in INDs and NDAs—which are often
followed by new research conducted to address knowledge gaps in key areas.
DARS scientists are currently involved in over 30 projects that advance drug development and safety
assessment to help inform regulatory decision making. Some highlights are presented below.
• Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis (SJS/TEN) are serious and potentially
life-threatening skin reactions that can occur in individuals treated with a variety of drugs, and are
therefore of significant concern to CDER and the larger medical community. To better understand
underlying mechanisms, DARS researchers conducted analyses that identified a specific protein as
a significant mediator of SJS/TEN. Leveraging a genomics database, DARS found that the drug
carbamazepine disrupted several genes, including one associated with carbamazepine-induced SJS/TEN.
This information can be applied in future drug reviews as well as postmarket safety oversight.
• In another effort to investigate drug-induced cardiac toxicity, DARS is part of a research consortium
of worldwide regulatory agencies, industry, and academic centers evaluating the possibility of predicting
drug-induced cardiac toxicity using a computer model of the human ventricular (heart muscle) cell. This
project involves evaluating drugs and then simulating their effects on electrical activity in the heart under
a wide range of conditions. The study seeks to develop a new regulatory pathway that will change current
cardiovascular safety testing guidelines and potentially eliminate the need for certain kinds of clinical
testing during drug development.
• DARS is pursuing development of industry standards for biomarker discovery and evaluation, including
gene-based research methods and reference data sets, to develop consistent criteria to measure tissue
damage in pathology samples.
• DARS is evaluating digital tools that may help to predict and explain drug-related adverse events. These
tools can be used to generate possible explanations for why an adverse event might occur, and have the
potential to help modernize drug safety analyses, to improve product labeling, and guide development in the
pre-market phase as well as supporting pharmacovigilance in the postmarketing period.
• DARS is evaluating a software program that generates adverse event profiles based on a drug’s
molecular targets in the human body, to predict adverse events not previously seen in clinical trials and
therefore not included in the original product label. Predictions of drug-related adverse events have already
been made using this approach. Planning is underway to perform similar analyses on many more drugs now
under review.
DARS research is tightly linked to current and emerging drug product reviews and
is customized to answer specific questions. The value of DARS’ resident expertise to
CDER regulatory science is enhanced through targeted collaborations across FDA
and with industry and academia, providing additional opportunities to build on its past
research successes and improve the way drugs are developed and reviewed.
This effort cannot be done alone but rather requires partnerships between FDA and
Creating 21st Century multi-sector stakeholders sharing the common goal of promoting drug safety and
safety science requires effectiveness to protect and improve public health.
the necessary expertise
of internal and external The Critical Path Initiative (CPI), launched in 2004, is FDA’s keystone initiative to
partners and scientific forge stakeholder collaborations to tackle various drug safety and drug development
collaborators. issues.
These strategic partnerships help to enhance the efficiency and quality of medical
product development, evaluation, and manufacture through scientific advances—
the “critical path”. When a new or growing public health concern is identified
that cannot be effectively addressed by a single organization, CDER and other
organizational components in FDA, operating within the framework of the CPI,
can ask a non-profit stakeholder organization to serve as a neutral convener that can
then leverage the expertise of numerous other stakeholders through a public-private
partnership (PPP).
Through the expertise and resources of external scientists working in the PPP model, vital new information
is shared in a pre-competitive space that is defining the path forward for drug safety and drug development
science. Through the products of such partnerships, CDER is better positioned to ensure that new drugs
are safe and effective prior to marketing, to continue to assess and monitor safety once drugs are on the
market, and to help patients and health care providers make better decisions about the drugs they use.
CDER integrates information from these sources with its own evaluations into an overall system of
postmarket surveillance and risk assessment, and uses this aggregated information when a drug’s identified
risks indicate a need to undertake any of a number of actions. These actions can include providing
additional safety information to the public, updating drug labeling, requiring postmarket studies or trials or
other risk management interventions, and, rarely, withdrawing approval of a drug.
When there is an unmet need for the treatment of a serious medical condition, CDER may use one or more
of its expedited programs, including the “fast track” or “breakthrough therapy” designations, intended to
bring drugs to market and expedite their availability to patients. CDER is also responsible for monitoring
the safety of these marketed drugs and reporting on those efforts.
In December 2015, the Government Accountability Office (GAO) issued a report responding to a
Congressional request that GAO provide information about aspects of CDER’s postmarket monitoring
activities.
GAO was tasked with providing information on CDER’s expedited programs and its postmarket monitoring
of expedited and nonexpedited drugs. GAO’s report examined the number and types of requests for fast
track or breakthrough therapy designation that CDER received, and the number and types of approved drug
applications that used an expedited program, focusing on “the extent to which FDA’s data on tracked safety
issues and postmarket studies allowed the agency to meet its reporting and oversight responsibilities.”
GAO analyzed CDER data on requests for fast track or breakthrough therapy designation and approved
drug applications that used an expedited program between October 1, 2006 and December 31, 2014.
GAO also interviewed relevant CDER staff and reviewed CDER information on tracked safety issues and
postmarket studies, concluding that:
• “FDA lacks reliable, readily accessible data on tracked safety issues and postmarket studies needed to
meet certain postmarket safety reporting responsibilities and to conduct systematic oversight.”
• “While internal control standards for the Federal government specify that information should be
recorded in a form and within a time frame that enables staff to carry out their responsibilities, and that
relevant, reliable, and timely information should be available for external reporting purposes,” GAO
found that CDER’s “data on postmarket safety issues and studies were incomplete, outdated, contained
inaccuracies, and was stored in a manner that made routine, systematic analysis difficult.”
GAO acknowledged that CDER has supported efforts to shorten development and streamline review of drug
applications through expedited pathways, but noted problems with “efforts to oversee and track potential
safety issues and postmarket studies once those drugs are on the market.”
In responding to the GAO report, CDER clarified that, to be approved, all drugs must meet the same
standards for safety and effectiveness—whether or not a particular drug has been the subject of an expedited
review program. CDER also noted that, based on continuous improvement processes launched prior to
the initiation of the GAO study, the challenges related to administrative tracking of the two aspects of the
Center’s postmarket safety work noted above had already been recognized and remedies were underway.
Consistent with GAO’s recommendations, CDER is continuing to address those issues through ongoing
improvement efforts.
PMRs CDER’s most recent annual assessment found that most open PMRs and PMCs
CDER may require drug are progressing on schedule.
companies to conduct
postmarketing studies or clinical Tracked Safety Issues
trials to further characterize
TSIs are one of the many ways in which information about significant safety
known or potential serious risks
issues is shared within CDER and across the Agency, joining the larger system
of a drug. These studies and
of safety oversight programs and resources described in this report. At the time
clinical trials are referred to as
the GAO report issued, CDER had already noted that its administrative tracking
postmarketing requirements,
of TSIs needed re-evaluation and updating. The Center recognized that the
or PMRs.
overall number of TSIs entered into the system established for tracking safety
PMCs issues was low compared to the number of postmarketing safety issues already
Drug companies frequently identified and, therefore, TSIs were not being effectively recorded in the database
recognize the importance of in a timely manner. CDER addressed this issue and continues to do so: As of
certain other postmarketing June 30, 2016, 154 new TSIs have been created and 171 retroactive TSIs have
studies and, at FDA’s request, been added to the database. New TSIs are being entered into the tracking system
commit to conducting those immediately or shortly after they are identified.
studies. A postmarketing
study that is agreed to, but
Optimizing Drug Safety Oversight
not required, is called a
CDER needs every available tool in its efforts to maintain a robust, versatile,
postmarketing commitment,
and technologically optimized drug safety oversight program. In response to
or PMC.
the GAO report and based on CDER’s own program assessments, the Center
continues to implement new processes and procedures to correct data and
address gaps in information. CDER is also engaged in activities to increase and
streamline the capture of specific information into its database and is facilitating
additional analyses for oversight of postmarket studies and safety issues—
reflections of the Center’s commitment to completing implementation of these
and other new safety monitoring processes and procedures in a timely manner.
Support for better treatments for pain and for opioid overdose. Naloxone is a drug used to
counteract the effects of opioid overdose. CDER recently approved naloxone in an intranasal form
(administered by spraying into the nose). Building on this approval, CDER is reviewing options,
including over-the-counter availability, to make naloxone more available. While CDER continues its
assessment, the Agency actively supports the Centers for Disease Control and Prevention guidelines
for prescribing opioids for the treatment of pain, and will facilitate the development of evidence and
improved treatments to bring broader access to overdose treatment, safer prescribing and use of opioids,
and ultimately, new classes of pain medicines without the same risks as opioids.
Reassessment of the risk-benefit approval framework for opioid use. CDER received advice from
the Agency’s Science Board in March 2016, and has engaged the National Academies of Sciences,
Engineering, and Medicine on how to take into account our evolving understanding of the risks of
opioids—not only for patients but also the risks of misuse by other persons who obtain them. Both
activities will generate publicly available reports serving to incorporate the broader public health
impacts of opioid abuse in CDER’s drug product approval decisions.
• Developing, and providing doctors with, better information about the risks of opioids and how to
prescribe these drugs safely.
• Improving scientific evidence to guide the use of opioid medications, particularly in long-term use
settings, by strengthening requirements for drug companies to generate postmarket data on the long-term
impact of using extended-release/long-acting opioids.
As many as 1.5 million Americans are injured or killed each year by inappropriate use of FDA-regulated
drugs, including intentional or accidental misuse, or errors such as receiving the wrong drug or wrong
dose from a pharmacy or doctor. Many of these deaths or injuries are preventable.
As an external public outreach complement to Safety First, CDER instituted the Safe Use Initiative
(SUI) to develop solutions within the broader healthcare environment that go beyond standard
regulatory methods to enhance the safe and appropriate use of drugs for all Americans. SUI seeks to
reduce preventable harm from medications by developing, implementing, and evaluating cross-sector
interventions with partners committed to safe and appropriate medication use. As part of this long-
term initiative, SUI brings together various parts of the health care system to develop interventions that
measurably reduce preventable harm from medications.
Multi-state Prescription Drug Monitoring Program Surveillance Database. In 2015 the SUI team
partnered with the Bureau of Justice Assistance and Brandeis University to develop a surveillance
database using state Prescription Drug Monitoring Programs that will enable CDER to identify opioid
use patterns.
Development of a risk score for severe hypoglycemic events. SUI partnered with
Kaiser Permanente to provide health care providers with a practical way to identify
patients at high risk of severe hypoglycemia and guide shared decision-making
with respect to modifications of glucose-lowering therapy in out-patient clinical
settings. The prediction model has been completed and validation is underway.
Healthy People 2020 – Medical Product Safety Topic Area Working Group.
Healthy People provides science-based, 10-year national objectives for improving
The Safe Use Initiative works
the health of all Americans. SUI team members are leading the Medical Product
to support the safe use of
Safety Topic Area Working Group as part of the HHS Healthy People 2020
prescription medicines and
Initiative. This group is revising a developmental objective focused on reducing
spans federal partnerships,
deaths from the use of pain medicines.
public-private workgroups,
Standardizing the preparation of medications to reduce errors. With SUI collaborations throughout
support, the University of Michigan gathered information on current practices health communities including
involving preparation of oral liquid medications in pharmacies, and developed patients and patient
voluntary standard concentrations in compounding medications to reduce the risk caregivers, as well as funding
of errors when patients change pharmacies or transition their care. This study original research to better
ended May 2015 and received the annual Cheers Award from the Institute for understand the nature of
Safe Medication Practices, recognizing superlative standards of excellence in the prescription medicine misuse
prevention of medication errors and adverse drug events. One of the investigators and errors.
is now working on a national standardization effort with SUI and the Association
of Health-Systems Pharmacists.
SUI’s ongoing efforts, working across a spectrum of activities to support and facilitate
the safe use of prescription medicines, spans federal partnerships, public-private
workgroups, collaborations throughout health and medical communities outside of
government, the enlistment of experts beyond medical providers and administrators
to include insurers, patients, and patient caregivers, as well as the funding of original
research to better understand the nature of prescription medicine misuse and errors.
The quality of drug products is a pivotal aspect of drug safety, making product
quality oversight a vital asset in CDER’s drug safety portfolio. In facilitating
needed growth in CDER’s product quality programs, Center Director Janet
Woodcock spearheaded the stand-up of the new CDER Office of Pharmaceutical
Quality (OPQ) in January 2015.
These tenets establish “One Quality Voice” for patients, providers, and industry,
creating the “critical mass” that ultimately assures the availability of safe,
effective, high-quality medicines to the American public.
OPQ recently celebrated its first anniversary, marking several milestones and
achievements including:
CDER has responded to many serious adverse events, including infections and
deaths, associated with compounded drugs that were contaminated or otherwise
compounded improperly. The most serious in a long history of outbreaks associated
with contaminated compounded drugs was an outbreak of fungal meningitis in late
2012. Over 750 patients in 20 states were diagnosed with a fungal infection after
receiving injections of the contaminated drugs, and 64 patients in nine states died.
Since the 2012 fungal meningitis outbreak, FDA has identified insanitary conditions
at many of the compounding facilities it has inspected. Among many other insanitary
conditions that put patients at risk, examples include dog beds and dog hairs in close
proximity to a sterile compounding room, pharmacies with dead insects in ceilings,
renovations made without any evidence of controls to protect sterile drugs from
contamination, use of coffee filters to filter particulates, compounding by personnel
Compounded drugs serve with exposed skin during sterile production, toaster ovens used for sterilization, and
an important role for a kitchen dishwasher used to clean sterile compounding equipment and utensils.
patients whose medical
Congress enacted the Drug Quality and Security Act (DQSA) in 2013. The DQSA
needs cannot be met by
removed certain provisions from section 503A of the Federal Food, Drug, and
an FDA-approved drug
Cosmetic Act (FDCA) that were found to be unconstitutional and created a new
product—such as a patient
section, 503B, in the FDCA. Under this new section, a compounder can become
who needs a medication
an “outsourcing facility.” Outsourcing facilities must comply with current good
to be made without a
manufacturing practice requirements, are inspected by FDA according to a risk-
certain dye to avoid an
based schedule, and must meet certain conditions, including reporting adverse events
allergic reaction. But drugs
to FDA associated with their compounded drugs.
that are compounded
improperly, or that are CDER has worked quickly to implement the compounding provisions of the DQSA,
compounded under while directing inspection and enforcement initiatives to protect the public from
substandard conditions, poor quality compounded drugs. In 2015 and so far in 2016, CDER:
can pose serious health
risks. • Continued inspection and enforcement efforts, as well as policy development,
related to human drug compounding.
• Issued more than 35 warning letters to compounders. In addition, in 2015, two compounders entered into
consent decrees of permanent injunction (a legal agreement to permanently comply with FDA regulatory
policies).
CDER has also continued to make significant progress in implementing the compounding provisions of the
FDCA by publishing policy documents applicable to compounding pharmacies and outsourcing facilities. In
2015 and so far in 2016, CDER has published twelve draft guidance documents, four final guidances, and a
draft memorandum of understanding with the states, and also held the first five meetings of the reconstituted
Pharmacy Compounding Advisory Committee.
As problems are identified at compounding pharmacies and outsourcing facilities across the country, CDER will
continue to vigorously pursue inspection and enforcement efforts.
FDA has been actively engaged in efforts to improve the security of the drug supply chain for many years
to protect patients from unsafe, ineffective, and poor quality drugs. Since the formation of the first FDA
Counterfeit Drug Task Force in 2003, the Agency has advocated for a multi-tiered approach to securing the
supply chain and protecting consumers from the threats posed by counterfeit or compromised drug products.
The ability to trace prescription drugs also plays a significant role in providing transparency and accountability
in the drug supply chain.
The Drug Supply Chain Security Act (DSCSA) was signed into law in 2013, providing for a uniform Federal
framework to be in place by 2023. This framework will identify and trace certain prescription drugs by outlining
critical steps to build an enhanced electronic, interoperable system to trace drug products as they are distributed
within the United States. This critically important system expands and improves FDA’s ability to protect the
public from exposure to drugs that may be counterfeit, stolen, tainted, or otherwise potentially harmful.
Since January 1, 2015, the law placed new requirements on drug manufacturers, repackagers, and wholesale
distributors, followed by dispensers (primarily pharmacies), to have systems and processes in place to
exchange product tracing information, conduct verification to identify suspect product, including steps such as
quarantining and investigating suspect product, notify FDA and trading partners when an illegitimate product is
found, and keep records.
FDA has issued several guidances for industry and continues to work with stakeholders on implementation
through public workshops and other meetings. To help inform the enhanced system of 2023, FDA is
coordinating with members of the supply chain and developing a pilot project program that will explore and
evaluate methods to enhance the safety and security of the pharmaceutical distribution supply chain. This pilot
project program will focus on utilization of the product identifier, which will enable product tracing down to the
level of an individual package.
As CDER’s preeminent resource for communicating human drug information, the Center’s Office
of Communications (OCOMM) serves at the nexus of virtually every drug safety issue, question,
or crisis that CDER faces. With nearly 100 staff members including health professionals,
communications specialists and web and graphic designers, OCOMM fulfills CDER’s internal
and external communication needs, including providing strategic communication advice to
Center and Agency leadership, developing and coordinating overarching public communication
initiatives and educational activities, and employing comprehensive communication approaches
to ensure consistent branding, messaging, and strategic direction of CDER’s communication
products—all critical aspects of drug product safety.
More than 65,000 requests for information are received annually, many of which involve some
aspect of drug safety, from how much or how little of a medicine to safely use, to safe prescribing
or dispensing practices, to responses on media-trending drug safety concerns. Queries arrive via
phone, electronic mail, and letter, from an array of stakeholders, including consumers, health care
professionals, academia, foundations and research organizations, and the pharmaceutical industry.
Safety inquiries can involve complex scientific or regulatory issues; in those cases OCOMM
coordinates extensive research and consultation needs with the subject matter experts needed to
craft accurate and balanced responses.
Another variation in public inquiry are national write-in campaigns, which are frequently
organized around a particular facet of medication safety. OCOMM organizes and manages
CDER’s response to these campaigns—in the last 18 months alone, OCOMM has sent over 2,000
emails and letters to 16 different campaigns.
Facebook. CDER became the first Center to engage the public on FDA’s Facebook
page, with 128 drug-related posts reaching over 4 million Facebook users.
Twitter. CDER became the first FDA Center with a Twitter account (@FDA_Drug_
Info). 616 Tweets were sent, and 40,764 new followers were gained over the last 18
months to reach a total of more than 200,000 Twitter followers. OCOMM organized
and led the first CDER Twitter Chat, #LungChat, which engaged major oncology
organizations, patients, advocates and health care professionals about lung cancer
treatments, and live-tweeted three of Center Director Janet Woodcock’s recent
Congressional testimonies.
Podcasts. FDA Drug Safety Podcasts provide Drug Safety Communications in audio
form and are available on iTunes and ReachMD radio—46 Drug Safety Podcasts
have seen 31,846 transcript visits and 135,388 audio downloads in 2015 and through
the first quarter of 2016.
• Celebrating 30 years at FDA. Dr. Woodcock reflects on breakthroughs in drug research and
regulation and makes some predictions for the future of drug products.
• Talking Translational Science. Dr. Woodcock defines translational science and how it can
positively affect drug development and review.
• Looking Back and Moving Forward in 2016. Dr. Woodcock discusses major events of 2015 and
priorities for 2016.
• Drug Compounding. Dr. Woodcock discusses drug compounding from both a safety and regulatory
standpoint.
• Working with Patient Advocacy Groups. Dr. Woodcock discusses regulatory guidances drafted and
submitted to FDA by patient advocacy groups.
Video. FDA Drug Info Rounds Videos target pharmacists ond other health care providers and are
promoted to State Boards of Pharmacy, pharmacy schools, and posted on YouTube. Many of the Drug
Info Rounds Videos produced in 2015 and 2016 focus on aspects of drug safety.
• REMS@FDA Tutorial presents the new and improved REMS website called REMS@FDA.
• Emergency Preparedness – Keeping Medications Safe discusses the importance of having a plan in
place for keeping medications safe as part of emergency preparedness.
• MedWatch Tips and Tools presents resources available to health care professionals to make reporting
to MedWatch easier than ever.
• Breakthrough Therapy discusses the breakthrough therapy designation, an exciting new program to
expedite drug development while maintaining safety oversight in the development process (Bronze Telly
Award).
• FAERS provides background information about the FDA Adverse Event Reporting System (FAERS)
database.
• REMS presents the many components of Risk Evaluation and Mitigation Strategies (REMS) and how
they can help manage a drug product with known or potential serious risks.
Drug Information Update. The FDA Drug Information Update email subscription service sends
bulletins to stakeholders with safety information about drug products including Drug Safety
Communications, drug approvals, CDER Statements, and industry guidances. The Drug Information
Update email service gained 28,999 new subscribers in 2015 (with a current total of 155,447), and the
service sent out 208 email messages.
DSCs are usually issued in conjunction with regulatory actions such as Safety
Labeling Changes for a number of reasons, including issues affecting a large Drug Safety Communications
number of patients, potentially serious or life-threatening adverse events, or are broadly circulated
medication errors that may result in serious or life-threatening adverse reactions. through a variety of channels,
including email newsletters,
The Drug Safety Communication web page is the most visited page on the FDA’s
podcasts, social media
web site. Forty-six DSCs were issued between January 1, 2015, and July 26, 2016,
and targeted outreach to
and during that time the DSC home page and the 46 individual DSC web pages
healthcare professionals,
received over a million page views. Outreach was significantly greater than that,
advocacy groups and other
however, as DSCs are much more broadly circulated through a variety of other
stakeholders.
channels, including several large listservs, email newsletters, podcasts, social
and traditional media, and targeted outreach to media, healthcare professionals,
advocacy groups and other stakeholders.
Although SLCs have been available online for many years, they were aggregated
and posted on a monthly basis. This meant that if a new safety concern was
reflected in an approved labeling change early in a month, that information was
not publicly posted until early in the following month—three to four weeks later.
As web and database technologies evolved, CDER recognized a need to apply
new digital functionalities that would not only shorten the time between an SLC’s
approval and when its safety information becomes available on the FDA’s website,
but to generally enhance an online visitor’s experience by improving the SLC web
platform’s clarity, access, and navigability.
The table below details the number of SLCs approved between January 2015 and
July 2016�more than 1,500 in all.
Boxed Warnings 87
Contraindications 83
Warnings 452
Precautions 548
TOTAL 1,676
In late 2015, a study of medical countermeasures messaging (launched in 2012) was completed,
offering insight and recommendations for developing effective communication initiatives for public
health emergencies. A second phase of this study will be undertaken in 2016 to test the various medical
countermeasures messages that were developed as a result of the initial study findings.
Other OCOMM risk communication research conducted in 2015 and through early 2016 included:
• A study to enhance FDA communications addressing opioids and other potentially addictive pain
medications, scheduled to be completed in 2017
• Research exploring issues of uncertainty and unintended consequences associated with prescription
drug communications
• A survey project building on prior focus group research that tests Drug Safety Communications
to better understand consumer recall, understanding of risk, and behavioral intentions related to DSC
content and formatting
As with all areas of science and technology, drug safety science is constantly evolving. CDER
recognizes that ongoing success in safety oversight and surveillance requires a continuing
ability to adapt to new information, new technologies, and new developments. CDER has
created a safety system that directs scientific expertise and rigor across the lifecycle of FDA-
approved drugs, building the Agency’s ability to navigate the inevitable changes to come
in medicine and health care.
Moving forward, all CDER’s safety efforts, as they remain thorough and systematic, will
continue to be designed to support parallel efforts to advance innovation and help ensure
that safe and effective new therapies are available to the American public as efficiently and
rapidly as possible.