Transcript
  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    1/147

    H E A L T H C A R E

    Drug Approval Trends at the FDA andEMEAProcess improvements, heightened scrutiny and industry

    response

    By Alison Sahoo

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    2/147

    Alison Sahoo

    Alison Sahoo is a pharmaceutical industry analyst with more than 10 years of

    experience researching the development of medicines and medical devices. She holds a

    B.S. in Physics from McGill University and an M.B.A. in International Business from

    Rutgers University.

    Copyright 2008 Business Insights LtdThis Management Report is published by Business Insights Ltd. All rights reserved.Reproduction or redistribution of this Management Report in any form for anypurpose is expressly prohibited without the prior consent of Business Insights Ltd.

    The views expressed in this Management Report are those of the publisher, not ofBusiness Insights. Business Insights Ltd accepts no liability for the accuracy orcompleteness of the information, advice or comment contained in this ManagementReport nor for any actions taken in reliance thereon.

    While information, advice or comment is believed to be correct at the time ofpublication, no responsibility can be accepted by Business Insights Ltd for itscompleteness or accuracy.

    ii

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    3/147

    Table of Contents

    Drug Approval Trends at the FDA, EMEA

    Executive Summary 10

    The shifting regulatory landscape 10

    Drug approval trends at FDA 11

    Drug approval trends at EMEA 12

    Industry response 13

    Global drug approval trends through 2012 14

    Chapter 1 The shifting regulatory landscape 18

    Summary 18

    Drug usage in the U.S. and Europe 19

    New molecular entities 20

    Generic drugs 21Rising usage of generics 21Generic drug application trends 22

    Indication expansions 23

    Drug approval issues 26

    Ensuring drug safety 27

    Safety shortfalls: rising adverse drug reactions 29Safety failures: drug withdrawals 30

    The importance of time to market 31Impact of cost constraints 34

    Conclusion 35

    Chapter 2 Drug approval trends at the FDA 38

    Summary 38

    Drug approval process 39Pressure to approve drug candidates 42

    ii i

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    4/147

    Future directions in the FDA drug approval process 43Ensuring drug safety 44Evolution of drug safety testing 44Pediatric safety testing 48

    Future directions in drug safety reviews 49Reducing time to market 50Fast track approvals 52Orphan drug approvals 53Decreasing FDA drug approvals 54

    Future directions in speeding drug approvals 56

    Delaying risky applications 58Rising data requirements 58

    Case study: hormone replacement therapy 59Case study: Galvus 60

    The future for risky drug applications 62

    Biosimilars 62

    The future of biosimilar approvals 62Canadian biosimilars legislation 63

    Rx-to-OTC switches 64New OTC drug categories 66

    Antihistamines 67Emergency contraceptives 69Hyperosmotic laxatives 71Ophthalmic allergy medications 72Proton pump inhibitors 73Weight loss medications 75

    Switch rejections 76Statins 77

    Future directions in nonprescription drug approvals 78Creation of a pharmacist dispensed class 79

    Marketing withdrawals 80

    Application withdrawals 81Future directions in marketing and application withdrawals 82

    Conclusion 82

    Chapter 3 Drug approval trends at theEMEA 86

    Summary 86

    Drug approval process 87History of European drug regulation 87The formation of the EMEA and the MRP 88

    The EMEA 89

    iv

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    5/147

    The Mutual Recognition Procedure (MRP) 91The National Competent Authorities 93The Common Technical Document 93Ensuring drug safety 96

    Creation of Scientific Advisory Groups 97Future directions in drug safety testing 98Reducing time to market 98Accelerated opinions 100Pediatric testing exemptions 101Decreasing EMEA approvals vs. FDA approvals 101Current EMEA approval levels 104Future directions in speeding drug approvals 104

    Delaying risky applications 105The future for risky drug applications 106

    Biosimilars 107New biosimilar guidelines 107Future directions in biosimilar approvals 108

    Rx-to-OTC switches 109Future directions in nonprescription drug approvals 110

    Marketing withdrawals 110

    Application withdrawals 110Future directions in marketing and application withdrawals 112

    Conclusion 112

    Chapter 4 Industry response 114

    Summary 114

    Background 115

    Improving safety testing 115Genomics 116Stem cell approaches 117Future drug safety testing improvements 118

    Raising investment in R&D 118Improving R&D productivity 119

    Contract research organizations 119Repositioning 121

    Future directions in R&D investment 122

    Public reporting of clinical study results 123United States 123Europe 124Association of the British Pharmaceutical Industry 125

    Future directions in clinical data reporting 126Strengthening regulatory processes 126

    Coalition for a Stronger FDA 126

    v

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    6/147

    Future directions in regulatory support 127

    Conclusion 128

    Chapter 5 Global drug approval trends to2012 130

    Summary 130

    Background 131

    Trends at the FDA 132Focus on drug safety 132

    New technology-based standards for drug application review 133

    Decreasing application review times 135Boosting integrity and accountability 136

    Trends at the EMEA 137Focus on drug safety 138Product Information Management (PIM) Project 139Cooperation with other regulators 140Incentives for small drug developers 140

    Convergence of regulatory approaches 141

    Orphan drug approvals 143

    Conclusion 144

    Chapter 6 Appendix 146

    Index 146

    List of FiguresFigure 1.1: Drug safety risk spectrum 28Figure 1.2: Growth in U.S. reported adverse drug reactions, 1995 - 2007 30Figure 1.3: U.S. R&D spending versus new drug approvals, 1995 2007 36Figure 2.4: U.S. drug approvals, 1995 - 2007 55Figure 3.5: Average number of days for centralized procedure positive opinions, 2004 - 2006 99Figure 3.6: Average number of days for orphan drug designation opinions, 2004 - 2006 100Figure 3.7: EMEA marketing withdrawals, 2000 - 2007 111

    vi

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    7/147

    List of TablesTable 1.1: Selected indication expansions for U.S. commercialized drugs, 2002 - 2007 25Table 1.2: Potential safety risks of popular drug classes 29Table 1.3: Significant drug discontinuations and affected patient groups, 1997 - 2007 33Table 2.4: Timeline of key FDA legislations 45Table 2.5: CDER Advisory Committees, 2008 48Table 2.6: U.S. New Molecular Entity (NME) Approvals, 2000 - 2007 56Table 2.7: Key U.S. Rx-to-OTC switches, 1997 - 2007 66Table 2.8: Key events in the approval of OTC nonsedating antihistamines 69Table 2.9: Recent withdrawals of U.S. marketing applications, 2002 - 2007 82Table 3.10: Leading European National Competent Authorities, 2008 94Table 4.11: U.S. Clinical trials and R&D spending, 2000 - 2007 118Table 4.12: Selected repositioning specialists 122

    vi i

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    8/147

    8

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    9/147

    Executive Summary

    9

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    10/147

    Executive Summary

    The shifting regulatory landscape

    In both the U.S. and Europe, the utilization of Rx medicines, including both generic

    drugs and new molecular entities, is rising strongly.

    Increasing use of medications carries safety risk. Inherent drug risks are generally

    related to toxicity and the pharmacokinetic properties of the drug itself.

    Drug safety risks run the gamut from minor discomforts such as the nausea

    associated with many classes of medications to life-threatening conditions like

    increased risk of liver toxicity caused by cholesterol-reducing statins.

    Over the past five years, the incidence of adverse drug reactions reported in both

    the U.S. and Europe has risen sharply.

    Marketing withdrawals may be required for drugs that have been associated with a

    significant level of extremely adverse effects. The most significant market

    withdrawal to date has been Mercks Vioxx, which was recalled in September 2004

    after more than 80m prescriptions had been written for the drug worldwide. The

    drug was linked to an increased risk of cardiovascular effects, with one study

    showing that patients taking Vioxx were twice as likely to have a heart attack as

    those taking naproxen.

    Mercks withdrawal of Vioxx has galvanized public scrutiny of drug approval

    processes, causing regulators, particularly the FDA, to exercise more caution in

    approvals.

    As more blockbuster drugs lose patent protection and R&D productivity continues

    to decline, minimizing time to market for new drugs is becoming increasingly

    important. However, approval delays can lengthen time to market.

    10

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    11/147

    While both the FDA and EMEA are attempting to reduce approval times, largely

    through expansion of fast track programs, delays remain common, as regulators

    attempt to verify the safety of increasingly complex products.

    Expanding R&D activities to ensure that compounds submitted for approval

    represent only the best possible candidates might improve approval rates and times,

    but the high and rising cost of R&D continues to constrain such expansion.

    Drug approval trends at FDA

    Although FDAs drug approval process has come under heavy scrutiny since the

    market withdrawal of Mercks Vioxx in 2004, allegations of undue pressure on

    reviewers to approve new medicines date to well before this time. The FDA is

    responding with a renewed effort to foster a culture of integrity from the top down.

    Incentives to increase the pediatric data available on drugs used by children have

    resulted in updated pediatric labeling for more than 120 drugs.

    Officially, the FDA has not changed its overall standards for drug approval in many

    years. On case by case basis, however, the agency appears to be requiring more

    data to verify safety.

    To provide speedier approval for certain drugs that address unique needs, the FDA

    has established three categories within which applications will be reviewed on a

    shorter timetable: Fast Track, Accelerated Approval and Priority Review.

    Under the Orphan Drug Act, nearly 1,700 medicines have been designated orphan

    drugs, and 249 have been approved for use.

    Despite ongoing attempts by FDA to increase the availability of medicines, there

    has been no real improvement in overall drug approvals since 1995.

    11

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    12/147

    While the number of clinical trials conducted in the U.S. continues to rise, the

    number of new molecular entities approved each year has declined since 2004.

    FDA is attempting to reduce approval times for generic medicines and is

    increasingly switching Rx products from classes not previously available over-the-

    counter to nonprescription use.

    FDA is asking for more data pursuant to applications for drugs that pose heightened

    safety concerns.

    While the EMEA has recently established an approval pathway that clarifies the

    regulatory process by which biosimilars can be approved, the FDA has not yet

    established such a process.

    Drug approval trends at EMEA

    The EMEA is currently putting systems in place for a more effective centralized

    procedure with a role comparable to that of the U.S. FDA. However, budgetary

    concerns will constrain development.

    In Europe as in the U.S., increasing reports of adverse drug reactions, expanding

    usage of products obtained over the Internet and associated issues with

    counterfeiting are creating imperatives for heightened drug safety testing.

    To provide greater oversight for all products during both the approval and post-

    marketing periods, the EMEA created a set of Scientific Advisory Groups in 2006.

    The EMEA has not recently made substantive official changes to its overall

    standards for drug approval. On case by case basis, however, European regulators

    appear to be exercising more caution in granting approvals, particularly requiring

    sufficient data to confidently verify drug safety.

    12

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    13/147

    In an effort to bring needed medicines to market more quickly, the EMEA has been

    attempting to reduce the time it spends reviewing marketing applications. After a

    slight increase in overall review time in 2005, review times were significantly

    reduced in 2006 for centralized procedures.

    In July 2006, the EMEA implemented a new review system to speed the approval

    of innovative drugs that respond to unmet medical needs or constitute a significant

    improvement over available methods of prevention, diagnosis or treatment.

    To streamline drug testing and eliminate the collection of unnecessary clinical data,

    the EMEA has relaxed requirements for pediatric trials for certain products.

    Despite improvements in processing speed and volume, the EMEA is reportedly

    approving fewer products than the FDA.

    In its approach to biosimilars, the EMEA has a significantly more comprehensive

    approval structure in place than the FDA and in fact, Europe is quickly becoming

    the global center for the development and production of biosimilar medicines.

    Industry response

    Pharmaceutical and biotech companies continue to work aggressively, both

    separately and together, to improve drug development processes.

    Several new technologies are emerging to help identify toxicity early in drug

    candidates, including genomics and stem cell approaches.

    One of the simplest, albeit least desirable, means to address rising data requests

    from regulators and increasing standards for approval is for drug makers to raise

    their investments in R&D. This has, in fact, occurred, with the number of U.S.

    clinical trials expanding by 6.9% per year from 2000 to 2007 and U.S. R&D

    spending increasing by 13.2% annually.

    13

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    14/147

    One of the most significant ways in which drug makers have, and continue to,

    achieve R&D productivity improvements has been through the utilization of CROs.

    Overall, CROs are able to shorten clinical testing times by as much as 30%, thereby

    leading to faster drug approvals. This has led to rising usage of CRO services, with

    global CRO revenues estimated at $16b in 2007 and continuing to grow by roughly

    10% per year.

    Over the past two years, the pharmaceutical industry has embraced repositioning,

    a systematic new means to analyze an entire library of failed experimental

    compounds to identify new therapeutic applications.

    Industry groups are seeking greater transparency of clinical trial results in an effort

    to self-police the industry and thereby proactively prevent potentially more

    restrictive requirements from the FDA or Congress. In both the U.S. and EU, new

    clinical trial databases have been established to provide details of drug testing

    results.

    On the theory that a stronger regulator can more effectively, and more quickly,

    process drug approvals, industry groups are banding together to call for increasedfunding for regulators.

    Global drug approval trends through 2012

    In recent years, drug safety regulators worldwide have become more cautious of

    new products in response to heightened public scrutiny. This trend is expected to

    continue through the foreseeable future.

    The FDAs Strategic Action Plan specifically notes that protecting U.S. consumers

    from unsafe medical products is its first and foremost responsibility, implying

    that speeding drug approvals is of secondary importance.

    A key component of the FDAs approach to ensuring drug safety through the

    approval process is likely to be increased requests for additional data to supplement

    marketing applications as well as post-marketing data.

    14

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    15/147

    New molecular entities (NMEs) are expected to be a key focus of the FDAs

    heightened interest in safety.

    The FDA is attempting to reduce its approval times through innovative new

    technology and training initiatives.

    While drug safety is also a key concern for the EMEA, the agency is also focused

    on expanding access to drugs and innovation.

    In 2006, the first marketing authorization application using the EMEAs new

    electronic PIM system was submitted.

    The EMEA has recently implemented legislation aimed at promoting innovation

    and development of new medicinal products by small and medium sized enterprises

    (SMEs).

    As globalization continues throughout the pharmaceutical, healthcare and other

    industries, the FDA and EMEA are increasingly sharing information about new

    drug candidates and adopting similar approaches to address issues that jointly affect

    their respective constituents.

    15

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    16/147

    16

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    17/147

    CHAPTER 1

    The shifting regulatorylandscape

    17

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    18/147

    Chapter 1 The shifting regulatorylandscape

    Summary

    In both the U.S. and Europe, the utilization of Rx medicines, including both

    generic drugs and new molecular entities, is rising strongly.

    Increasing use of medications carries safety risk. Inherent drug risks are generally

    related to toxicity and the pharmacokinetic properties of the drug itself. Drug safety risks run the gamut from minor discomforts such as the nausea

    associated with many classes of medications to life-threatening conditions like

    increased risk of liver toxicity caused by cholesterol-reducing statins.

    Over the past five years, the incidence of adverse drug reactions reported in both

    the U.S. and Europe has risen sharply.

    Marketing withdrawals may be required for drugs that have been associated with

    a significant level of extremely adverse effects. The most significant market

    withdrawal to date has been Mercks Vioxx, which was recalled in September

    2004 after more than 80m prescriptions had been written for the drug worldwide.

    One study showed that patients taking Vioxx were twice as likely to have a heart

    attack as those taking naproxen.

    Mercks withdrawal has galvanized public scrutiny of drug approval processes,

    causing regulators, particularly the FDA, to exercise more caution in approvals.

    As more blockbuster drugs lose patent protection and R&D productivity

    continues to decline, minimizing time to market for new drugs is becomingincreasingly important. However, approval delays can lengthen time to market.

    While both the FDA and EMEA are attempting to reduce approval times, largely

    through expansion of fast track programs, delays remain common, as regulators

    attempt to verify the safety of increasingly complex products.

    Expanding R&D activities to ensure that compounds submitted for approval

    represent only the best possible candidates might improve approval rates and

    times, but the high and rising cost of R&D continues to constrain such expansion.

    18

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    19/147

    Drug usage in the U.S. and Europe

    As the populations of the U.S. and Europe continue to age, their utilization ofprescription medicines is rising. While persons in their 20s use just two prescriptions

    per year, on average, adults ages 65 to 69 have 14 prescriptions filled per year, and

    those ages 80 to 84 have about 18 prescriptions filled per year, according to U.S.

    Pharmacist. These medicines address a variety of conditions, many of which are age-

    related, including high cholesterol, hypertension, arthritis, insomnia, etc. At the same

    time, obesity continues to rise with approximately two thirds of the U.S. and European

    populations overweight in early 2008 and one third obese. As excess weight is also

    associated with a range of medical conditions including cardiovascular issues, diabetes,

    gastrointestinal issues, etc., prescription drug usage in this population is similarly

    rising. The proliferation in lifestyle drugs, such as erectile dysfunction medicines,

    smoking cessation aids, etc., has further fueled the boom in prescription drug usage. By

    the end of 2007, average U.S. per capita usage of prescription drugs had risen to more

    than 13 prescriptions annually, compared with about 7 in 1992. Trends in Europe are

    similar.

    Together, the U.S. and Europe jointly comprise roughly 75% of the total global Rx

    drug market. In 2007, total market sales reached roughly $690m, up 6.5% from the

    prior year. In 2008, sales are expected to slow slightly but nonetheless post healthy

    gains of 5% to 6%. This strong and continued expansion in the usage of prescription

    medicines has given new importance to the processes by which drug candidates are

    reviewed and approved for use.

    In general, regulators in the U.S. and Europe are charged with the review of three types

    of prescription drug products:

    New molecular entities;

    generic drugs, which are based bioequivalent to previously approved products;

    previously approved drugs seeking expansions of their original indications.

    19

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    20/147

    As discussed in the subsections below, each of these drug candidates poses different

    types of challenges and is therefore subject to different approval trends. Because the

    greatest gains are to be earned from new molecular entities, and therefore most drug

    spending is focused on novel compounds, this report will center on approval trends for

    these products with only a peripheral discussion of generic approvals and indication

    expansions.

    New molecular entities

    New molecular entities represent novel compounds that are submitted to regulators for

    the first time. In the U.S., drug makers file a New Drug Application (NDA) with the

    FDA, while in Europe, applicants file similar documents either with individual national

    regulators or centrally through the mutual recognition procedure (MRP). In all cases,

    the goal of the review process is to provide sufficient information to establish that:

    The drug is safe and effective for its proposed usage, with benefits outweighing

    risk;

    the drugs proposed labeling is appropriate;

    the methods used to manufacture the drug and maintain quality are adequate.

    This process is particularly important since new molecular entities increasingly address

    complicated conditions with highly sophisticated science. Applications typically

    contain tens, and sometimes hundreds, of thousands of pages and detail the results of

    animal studies, human clinical trials, drug formulation, human drug metabolism,

    manufacturing, processing and packaging. Biologic products, such as vaccines and

    many recombinant proteins, are approved by FDA via a Biologic License Application

    (BLA), rather than an NDA. Manufacture of biologics is considered to differ

    fundamentally from that of less complicated chemicals, requiring a different approval

    process.

    20

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    21/147

    Generic drugs

    Generic drugs are essentially copies of products that have previously been approved.

    They are generally introduced when the patent on the original drug expires or is found

    to be invalid. Generics may be introduced by the developer of the original drug, but are

    more often launched by specialty generics manufacturers as a means to enter a new

    market.

    Rising usage of generics

    Because generic drug makers need not reproduce safety studies when seeking approval

    for product that is bioequivalent to an approved medicine, their R&D costs are

    significantly lower and they can therefore introduce products at steep discount to

    original drugs. According to the nonprofit Kaiser Family Foundation, for example, in

    2006 the average U.S. brand-name prescription cost more than three times the average

    generic, at $111 compared with $32.

    As health care costs continue to rise, this significant price advantage has resulted in

    dramatic growth for the generic industry as generic drug makers aggressively challenge

    patents in order to obtain rights to ever more top-selling medicines. In 2008, the global

    generic drug market is expected to reach about $80b. Growth is particularly strong in

    the U.S., where health care is funded primarily through the private sector. In 2007,

    generics accounted for more than half of all prescriptions filled, up from 44.9% in

    2003. Virtually all health care plan providers are attempting to contain spending on

    prescription drugs, implementing incentives for plan participants to use generics rather

    than branded products. This may take the form of step therapy (sequential use of least

    expensive medications before more expensive drugs), as a formulary alternative for

    which plan participants make the lowest co-payment or in some cases, as the only

    medicine in a given therapeutic category that the plan will cover.

    21

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    22/147

    Generic drug application trends

    As a result of this rising demand for low cost medicines, generic approval rates

    continue to rise. In its 2007 fiscal year, the U.S. FDA approved 683 generic drugs, a

    30% increase from its 2006 fiscal year. The agency has said that during 2008, it will

    bolster its generic drug program so that it can more quickly process the rising number

    of generic drug applications it expects to receive. These Abbreviated New Drug

    Applications (ANDAs) may be filed for any molecular entity except biologics, such as

    recombinant proteins.

    In Europe, health care is largely government funded but the level of generic drug usagevaries considerably with the regulations of each country. Nonetheless, usage of

    generics has been rising due to initiatives in France, U.K. and Germany to reduce

    government drug spending. This includes, for example, a partial reference pricing

    system established in France in 2003 under which branded products off patent are only

    reimbursed at the level of the generics, forcing patients to pay the difference for the

    branded version. In Germany, a 2003 mandate for aut idem prescription writing

    required pharmacists to fill prescriptions with one of the five cheapest generic versions

    for all drugs off patent. Branded products can only be used if the physician specifically

    indicates not to use a generic. This has resulted in generics accounting for about 40%

    market share by volume within a year of launch and nearly 70% within three years.

    Over the next five years, more than two dozen drugs with global sales of $500m or

    more will lose patent protection, creating significant new product opportunities for

    generics manufacturers, and stimulating further growth in generics usage. Key products

    that will lose patent protection in one or more major markets worldwide in 2008

    include Mercks Fosamax osteoporosis medication, Johnson & Johnsons Risperdal for

    schitzophrenia and Topamax for migraine, GlaxoSmithKlines Lamictal antiepileptic

    and Abbott Laboratories Depakote for the treatment of bipolar disorder.

    22

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    23/147

    Indication expansions

    An indication expansion broadens a drugs usage beyond its original intended use. Theexpanded usage may take a variety of forms, including:

    Expanded usage of the drug under more flexible conditions, such as for

    monotherapy instead of therapy in combination with another drug;

    extension of the drugs original usage to new patient populations, like children

    and/or adolescents;

    extension of the drugs usage to related indications, such as the approval of an

    antibiotic for specific types of infections;

    addition of entirely new therapeutic indications, such as treatment of additional

    diseases or conditions.

    While all of these extensions serve to increase the size of the potential patient

    population for the drug, pediatric expansion and particularly the addition of large new

    therapeutic indications generally have the most significant effect upon the drugs

    usage. They can also serve as an effective means of delaying generic competition, since

    regulations in both the U.S. and the E.U. provide some additional protection for drugs

    whose usage is extended.

    In the U.S., innovator companies are awarded three years of data exclusivity for a

    change to a products label that requires clinical trials to be conducted, although

    changes to the Medicare Modernization Act of 2003 have limited the opportunity to

    generate multiple 30-month stays. Originator companies had historically relied upon

    listing patents covering new indications in the Orange Book, thereby forcing generics

    manufacturers to make a paragraph IV certification in its Abbreviated New Drug

    Application (ANDA), and generating an automatic 30-month stay of approval.

    In Europe, manufacturers have long taken advantage of the requirement for generics

    companies to use the Mutual Recognition Procedure (MRP) process but also have the

    same Summary of Product Characteristics (SmPC) as the listed drug in each market, to

    23

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    24/147

    24

    extend patent protection by filing different indications in different markets. This results

    in SmPCs for the listed drug differing between regions and makes it impossible for a

    generic to gain approval for the full range of indications in each market via the MRP. A

    related action that can delay generic competition, even if SmPCs are harmonized across

    markets, is the filing of patents for new indications, which may expire at different times

    in different countries. Thus the generic company would be prevented from marketing

    the product for a particular indication. This can prevent launch of the product even for

    patent expired indications. The mention of a patented indication in the generics

    companys marketing authorization (MA) amounts to patent infringement, but

    excluding the patented indication can result in difficulties in gaining approval since the

    generic SmPC would differ from that of the originator product.

    Because of these benefits, indication expansion is a common lifecycle management

    strategy, with more than four fifths of the 50 top selling brands in 2007 having had

    additional indications approved since their initial U.S launch. Developers of

    blockbuster drugs often investigate additional indications particularly usage

    expansion to younger patients as a means to extend the products patent life and

    delay generic competition. This can give rise to multiple new indications for a single

    drug, as shown in Table 1.1.

    Often, a manufacturer will obtain information from external sources suggesting a

    potential new indication. This may arise from physicians, researchers and others who

    notice additional therapeutic benefits from a particular product, as well as similar

    competing products that obtain approval for additional indications. When this latter

    event occurs, it is not unusual for all the drugs within a particular class to one by one

    undergo an indication expansion. This occurred, for example, with Abbotts, Amgens

    and Johnson & Johnsons biological response modifiers for treatment of rheumatoid

    arthritis. Infectious disease, central nervous system disorders and cardiovascular

    disease remain popular areas for new indication expansion, largely due to the high

    numbers of patient sub-populations in these areas and the high commercial value of

    related products.

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    25/147

    25

    Table 1.1: Selected indication expansions for U.S. commercialized drugs, 20

    Company Drug Original Indication Expanded Indicatio

    Pfizer Lyrica Epileptic seizures Fibromyalgia Pfizer Celebrex Osteoarthritis Juvenile rheumatoid Johnson & Johnson Remicade Rheumatoid arthritis Chronic severe plaquSchering-Plough Avelox Infections Intra-abdominal infeWyeth Effexor Depression Panic attacks Abbott Humira Rheumatoid arthritis Psoriasis Johnson & Johnson Remicade Rheumatoid arthritis Ulcerative colitis Johnson & Johnson Levaquin Infections Bacterial sinusitis Johnson & Johnson Remicade Rheumatoid arthritis Psoriasis Johnson & Johnson Remicade Rheumatoid arthritis Active ankylosing spRoche Xenical Obesity Delay onset of type 2

    in obese patients with

    Johnson & Johnson Topamax Epileptic seizures Migraines Amgen Enbrel Rheumatoid arthritis Psoriasis Roche Xenical Obesity; patients to age 16 Patients to age 12 Shire Adderall ADHD; pediatric patients ADHD; adults

    Novartis Lescol Cholesterol reduction Risk reduction in correvascularization in pcoronary heart diseas

    Merck Singulair Asthma Allergies Pfizer Zyrtec Allergies; patients to age 2 Allergies; patients toGlaxoSmithKline Flonase Allergies Non-allergic nasal syAllergan Botox Various medical indications Cosmetic/aesthetic uTAP Pharmaceuticals Prevacid Ulcers and heartburn; to age 11 Patients to age 1

    Source: Company news releases and public filings

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    26/147

    Expansion of a drugs usage to pediatric patients is not appropriate for all products,

    however, since many of the most widely-used drugs address conditions that are most

    prevalent among older persons. These include medications that lower cholesterol,

    control hypertension, address various cardiovascular conditions, mitigate depression,

    etc.

    Some developers employ extensive indication expansion strategies, particularly for

    products whose usage can be leveraged to address closely related conditions. For

    example, through the end of 2007, Johnson & Johnson had obtained nine U.S.

    approvals for new indications for its Remicade, a biological response modifier initially

    cleared to market for the treatment of rheumatoid arthritis in 1998 and Allergans

    Botox (botulinum toxin) had been cleared for more than 20 different indications related

    to muscle-freezing in markets around the world.

    Drug approval issues

    With the continuing introduction of newer and better drugs that address a broader range

    of life-threatening and debilitating conditions, worldwide use of prescription medicines

    has soared over the past decade. This has resulted in significant economic and quality

    of life benefits. For example, the Organization for Economic Cooperation and

    Development estimates that over the past 40 years, new medicines have halved the

    number of hospital admissions for a variety of serious conditions including mental

    illness, infectious diseases and ulcers. As this has occurred, early infancy diseases have

    declined by 80% worldwide, ischemic heart disease has fallen by 68% and

    hypertensive heart disease has decreased by 67%. This has a direct dollar benefit, since

    for every $1 spent on pharmaceuticals, an estimated $4 is saved on hospital expenses

    and other therapeutic procedures.

    However, increasing use of medications also carries safety risk. Each year, about

    500,000 persons in the U.S. and Europe die from prescription drug reactions. Drug

    26

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    27/147

    safety issues have particularly come into the limelight with the market withdrawals of

    several COX-2 inhibitors on new safety information. However, while more

    comprehensive drug testing might identify potential problems in new products before

    they come to market, such testing will also increase time to market and development

    cost. These problems directly impact both drug makers as well as consumers.

    Ensuring drug safety

    Since the penning of the Hippocratic Oath admonishing doctors to do no harm, the

    medical community has long recognized the need to ensure that therapies intended to

    help patients do not, in the process, cause them more harm than good. In terms of drug

    safety, this imperative results in a continued need to balance access to new life-saving

    or life-enhancing therapies with potential safety risks that could result from adverse

    reactions. Although these risks may also arise from physician prescribing errors,

    pharmacist dispensing errors and/or inadequate patient compliance, this report will

    focus on inherent drug safety risks.

    Inherent drug risks are generally related to toxicity and the pharmacokinetic properties

    of the drug itself. While they can often be managed with appropriate labeling, dosing

    and monitoring of use, drugs that demonstrate an unacceptably high risk of adverse

    reactions which are believed to be generally unmanageable are disqualified from use.

    This usually occurs as soon as the risk is discovered and quantified, preferably early in

    the development process before more expensive clinical testing has begun.

    Manufacturers generally try to fail early, fail cheap by identifying significant safety

    risks early in the testing process. In some cases, the drug is eliminated from

    consideration by regulators during their review process and in a relatively small

    number of instances, a drug is removed from the market after it has been launched as

    new information about its effects come to light. This may occur either in the form of a

    voluntary recall by a manufacturer, as when Merck discontinued Vioxx in late 2004, or

    upon request by regulators, as FDA subsequently required of Bextra.

    27

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    28/147

    Medications with identified but manageable risks, which comprise the bulk of

    prescription drugs sold today, may be approved with expanded labeling requirements

    advising patients and healthcare providers of potential safety issues. These may contain

    contraindications for use applying to certain patient populations or may require regular

    physician monitoring of use. The small number of drugs that have not been associated

    with significant or severe side effects are generally not subject to these restrictions.

    Figure 1.1shows the spectrum of safety risk along which each drug sits and resulting

    restrictions on its use.

    Figure 1.1: Drug safety risk spectrum

    Generally unsafe

    under any

    circumstances

    Safe under certain

    conditions

    Generally safe for a

    wide population

    Few restrictions

    Labeling restrictsusage; manufacturer

    may recommend

    monitoring

    Manufacturerdiscontinues testing

    or drug not approved

    or drug recalled

    Increasing safety

    Generally unsafe

    under any

    circumstances

    Safe under certain

    conditions

    Generally safe for a

    wide population

    Few restrictions

    Labeling restrictsusage; manufacturer

    may recommend

    monitoring

    Manufacturerdiscontinues testing

    or drug not approved

    or drug recalled

    Increasing safety

    Source: Authors analysis Business Insights Ltd

    These safety risks run the gamut from minor discomforts such as the nausea and

    headaches associated with many classes of medications to life-threatening conditions

    like increased risk of liver toxicity caused by cholesterol-reducing statins. Although in

    most cases, the likelihood of serious side effects is quite small, for some groups of

    drugs, the risk is higher. These include some very broadly used classes of medications,

    such as those that address pain relief, asthma, migraine, osteoporosis, hypertension and

    other widespread conditions. Table 1.2 lists some widely used groups of medications

    and side effects that have been reported in clinical and post-marketing experience.

    28

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    29/147

    Table 1.2: Potential safety risks of popular drug classes

    Drug Class Indication Potential Reactions Safeguard

    COX-2 inhibitors Pain relief Risk of stroke, heart attack Expanded labeling,recalls

    Statins Cholesterol Liver toxicity Liver testsreduction

    NSAIDs Pain relief Ulcers Expanded labeling,contraindications

    Bisphosphonates Osteoporosis Gastrointestinal effects Expanded labeling,contraindications

    Hormone replacement Symptoms of Cardiovascular effects Expanded labeling,therapy menopause contraindications

    Leukotriene receptor Asthma Liver toxicity Expanded labeling,antagonists contraindications,liver

    tests

    Triptans Migraine Cardiovascular effects Expanded labeling,contraindications

    ACE inhibitors Hypertension Renal impairment Expanded labeling,physician supervision

    Source: Authors analysis Business Insights Ltd

    Safety shortfalls: rising adverse drug reactions

    In 2007, the estimated number of adverse drug reactions reported to the FDA from

    manufacturers and health care professionals exceeded 500,000, compared with

    approximately 150,000 in 1995. As shown in Figure 1.2, this 10.4% annual growth

    demonstrates a trend of increasing incidence of dangerous drug side effects.

    Although prescription volume has risen over the same period of time, growing from

    2.2b prescriptions in 1995 to 3.8b in 2007, adverse reactions are occurring in an

    29

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    30/147

    increasingly larger portion. While 0.007% of all prescriptions produced an adverse

    reaction in 1995, by 2007 this proportion had almost doubled to an estimated 0.013%.

    Affected populations increasingly include vulnerable groups such as seniors, which are

    expanding with the aging of the Baby Boomers.

    Figure 1.2: Growth in U.S. reported adverse drug reactions, 1995 - 2007

    0

    100,000

    200,000

    300,000

    400,000

    500,000

    600,000

    1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007

    ReportedAdverseDrugEvent

    Source: FDA Business Insights Ltd

    Safety failures: drug withdrawals

    Drug recalls are intended to prevent unsafe drugs from use and may be either

    temporary or permanent. Temporary recalls may involve one or a small number of

    batches of a particular drug that experience problems in distribution or manufacturing.

    These may include a variety of issues such as subpotency, stability data that does not

    support a drugs expiration date, dissolution failure, label mix-ups, the presence of a

    foreign substance in the product, pH failures or contamination of non-sterile products.

    30

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    31/147

    The vast majority of recalls represent temporary suspensions, with only a small

    proportion comprising permanent market withdrawals.

    While the number of significant safety-related drug withdrawals is relatively low, at

    less than 3% of approved Rx products, many have been very high profile, representing

    a large number of users and a high number of associated fatalities or other significant

    events. For example, Bayers cholesterol reducing statin, Baycol, was initially

    approved in the U.S. in 1997, then recalled in August 2001 following the deaths of 52

    patients worldwide. The deaths had been caused by rhabdomyolysis, a muscle ailment

    that had been known to be a possible side effect of all statin drugs, however, its

    incidence had been much higher and more serious among Baycol patients than among

    other statin users. Between 1997 and 2001 the drug was prescribed for more than 6m

    patients worldwide.

    Table 1.3shows significant Rx drug discontinuations that have occurred since 1997. In

    general, most of these withdrawals occurred on a worldwide basis, with a withdrawal in

    one market followed by withdrawals in other global markets. The most significant

    market withdrawal to date has been Mercks Vioxx, which was recalled in September

    2004 after more than 80m prescriptions had been written for the drug worldwide since

    its introduction in 1999. The drug had been linked to an increased risk of

    cardiovascular effects, with one study showing that patients taking Vioxx were twice as

    likely to have a heart attack as those taking naproxen.

    The importance of time to market

    As more blockbuster drugs lose patent protection and R&D productivity continues to

    decline, minimizing time to market for new drugs is becoming increasingly important.

    As of early 2008, an unprecedented number of blockbuster drugs have lost patent

    protection and become subject to competition from low priced generics, slowing the

    double digit growth the pharmaceutical industry had experienced previously. From

    2000 to 2005, for example, the compound annual growth rate (CAGR) for global

    31

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    32/147

    32

    blockbuster revenues was almost 20% and comprised nearly a third of a typical

    blockbuster marketing companys total revenues, on average. For some companies,

    including Merck, Pfizer and Amgen, blockbusters comprise more than half of total

    sales. However, when low cost generic competition arises, sales of cannibalized drugs

    fall by up to 80%, putting substantial pressure on drug makers to replace this lost

    revenue with sales from new products.

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    33/147

    33

    Table 1.3: Significant drug discontinuations and affected patient groups, 19

    Drug Manufacturer Use Recall Date Reason

    Zelnorm Novartis IBS March 2007 Cardiovascular effects

    Bextra Pfizer Pain April 2005 Cardiovascular effects

    Vioxx Merck Pain September 2004 Cardiovascular effects

    Baycol Bayer Cholesterol August 2001 Fatal rhabdomyolysis

    Raplon Organon Pain March 2001 Deaths from bronchospasm

    Lotronex GlaxoSmithKline IBS November 2000 Ischemic colitis

    Propulsid Johnson & Johnson Heartburn March 2000 Fatal heart rhythm disturbances

    Rezulin Pfizer Diabetes March 2000 Fatal liver toxicity

    Raxar GlaxoSmithKline Infection October 1999 Fatal heart rhythm disturbances

    Duract Wyeth Pain June 1998 Fatal liver toxicity

    Posicor Roche Hypertension June 1998 Fatal drug interactions

    Redux Wyeth Weight loss September 1997 Fatal heart valve damage

    Source: Company news releases and public filings

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    34/147

    However, approval delays can lengthen time to market. For potential blockbuster

    drugs that address large markets of chronic disease sufferers such as high cholesterol,

    hypertension or diabetes patients, excess time spent in testing results in significant lost

    sales. This further exacerbates financial pressure, since each day that a blockbuster

    drug with sales of $1b per year is delayed to market costs nearly $3m in sales.

    Approval delays also extend the time consumers must wait for needed medicines. In

    the U.S. and Europe, 10 to 15 years are typically required to take a drug from the

    laboratory through regulatory approval. Up to 6 years of this may be spent in pre-

    clinical testing, another 2 years may be devoted to Phase I evaluation, 3 years may be

    needed for Phase II testing and an additional 4 years may be necessary for Phase III

    studies. Pre-clinical studies include toxicology and other safety studies, conducted both

    in vitro and in animal models. Phase I studies evaluate drug safety, determine safe

    dosage ranges and identify side effects in small human groups of 20 to 80 volunteers.

    Phase II tests measure effectiveness and further evaluate safety in large groups of 100

    to 300 patients. Phase III trials confirm effectiveness, monitor side effects and compare

    the drug to other commonly used treatments in large patient groups of 1,000 to 4,000 ormore individuals. During this process, many drug candidates are eliminated of as

    many as 10,000 initially screened compounds, 250 enter pre-clinical testing, 5 enter

    clinical evaluation and just one is finally approved.

    While both the FDA and EMEA are attempting to reduce approval times, largely

    through expansion of fast track programs, delays remain common, as regulators attempt

    to verify the safety of increasingly complex products. For example, while the FDA has

    decreased average approval time from 16.6 months in 1995 to 11.5 months in 2006,

    approval times increased during four of these 11 years. These trends are examined in

    more detail in Chapters 2 and 3.

    Impact of cost constraints

    While expanding R&D activities to ensure that compounds submitted for approval

    represent only the best possible candidates might theoretically improve approval rates

    34

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    35/147

    and times, the high and rising cost of drug development continues to constrain such

    expansion.

    In 2007, drug developers worldwide spent an estimated $120b on drug development,

    up from about $2b in 1980. This includes pre-clinical, Phase I, II and III clinical

    testing. While a new drug cost on average $138m to take from conception to FDA

    approval in 1975, the average development cost had risen to $1b by 2007, outpacing

    inflation by more than 250%. In the U.S. alone, R&D spending has risen from $4b in

    1985 to $55b in 2006, according to the Pharmaceutical Research and Manufacturers

    Association of America (PhRMA). This is the result of several factors including

    increasing regulatory requests for more study data, more complicated science and more

    complex disease targets.

    Such rising expenditure on R&D would not be as troublesome if the number of new

    drugs were rising. However, this is not the case. As shown in Figure 1.3, although

    R&D spending has consistently increased over the past decade, the number of new

    drug approvals continues to fluctuate with no apparent relationship to spending.

    Conclusion

    In both the U.S. and Europe, recent marketing withdrawals have led to a heightened

    focus on drug safety, which in turn has impacted drug approval numbers and times.

    Although these considerations are not new, the intensity of public scrutiny, to a certain

    extent, is. This has created a new operating environment, in which regulators

    increasingly weigh the risks of new product against its benefits. When these risks

    cannot be adequately understood, regulators do not hesitate to request more data to

    validate or refute conclusions. This, in turn, is contributing to a rise in the size and

    number of clinical trials as well as overall research budgets.

    35

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    36/147

    Figure 1.3: U.S. R&D spending versus new drug approvals, 1995 2007

    0

    20

    40

    60

    80

    100

    120

    140

    1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007

    Number

    0

    10

    20

    30

    40

    50

    60

    70

    $b

    Number of approvals R&D expenditure

    Source: FDA, PhRMA Business Insights Ltd

    36

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    37/147

    CHAPTER 2

    Drug approval trends at theFDA

    37

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    38/147

    Chapter 2 Drug approval trends at theFDA

    Summary

    Although FDAs drug approval process has come under heavy scrutiny since the

    market withdrawal of Mercks Vioxx in 2004, allegations of undue pressure on

    reviewers to approve new medicines date to well before this time. The FDA has

    renewed effort to foster a culture of integrity from the top down.

    Officially, FDA has not changed its standards for drug approval in many years.

    On case by case basis, however, FDA appears to require more data to verify

    safety.

    To provide speedier approval for certain drugs that address unique needs, the

    FDA has established three categories within which applications will be reviewed

    on a shorter timetable: Fast Track, Accelerated Approval and Priority Review.

    Incentives to increase the pediatric data available on drugs used by children have

    resulted in updated pediatric labeling for more than 120 drugs.

    Under the Orphan Drug Act, nearly 1,700 medicines have been designated orphan

    drugs, and 249 have been approved for use.

    Despite ongoing attempts by FDA to increase the availability of medicines, there

    has been no real improvement in overall drug approvals since 1995.

    While the number of clinical trials conducted in the U.S. continues to rise, the

    number of new molecular entities approved each year has declined since 2004.

    FDA is attempting to reduce approval times for generic medicines and isincreasingly switching Rx products from classes not previously available over-

    the-counter to nonprescription use.

    FDA is asking for more data pursuant to applications for drugs that pose

    heightened safety concerns.

    While the EMEA has established an approval pathway that clarifies the process

    by which biosimilars can be approved, the FDA has not yet established such a

    process.

    38

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    39/147

    Drug approval process

    The U.S. FDAs drug testing and approval process consists of a series of discrete andwell-defined stages, each of which represents an opportunity to disqualify a drug that

    exhibits an unacceptable safety profile. Because the expense of testing usually

    increases with each stage, developers usually seek to disqualify marginal or borderline

    candidates early to minimize spending on products that will fail later. These stages

    include:

    Submission of a Notice of Claimed Investigational Exemption for a New Drug

    (IND);

    pre-clinical testing;

    Phase I clinical testing;

    Phase II clinical testing;

    Phase III clinical testing;

    submission of a New Drug Application (NDA).

    The IND was established by the 1962 Kefauver-Harris Amendment to the Federal

    Food, Drugs & Cosmetics Act of 1938, and generally contains all of the scientific

    information known about the investigational drug to be studied. After an IND has been

    filed with FDA, the sponsor must wait 30 days while FDA reviews the submission. If

    the FDA has not contacted the sponsor with questions about the IND filing, the initial

    introduction study may begin on the 31st day. In some cases, the drug developer may

    request a waiver of this rule, such as for studies of a drug previously approved for a

    different indication in a new condition. Since the safety of the drug in humans had

    already been established, FDA may allow the new clinical investigations to proceed

    immediately, since only efficacy in the new condition must be established.

    39

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    40/147

    Prior to human clinical testing, pre-clinical testing of new drugs generally includes

    three types of studies, all of which are performed in animals:

    Pharmacology studies that include screening and comparative tests with other drugs

    of known potency or activity; these determine what pharmacologic properties a

    drug has and which body systems the drug will affect;

    toxicology studies;

    reproductive and teratology studies; these evaluate the effects of individual drugs

    on reproductive activity, fetal development, lactation and postnatal neonatal

    development in animals.

    Phase I clinical trials involve the administration of the investigational drug to normal,

    healthy male volunteers. Females are usually excluded from these trials due to the

    possible teratogenic effect of a drug. The purpose of the studies is to determine human

    tolerance to increasing single doses as well as multiple doses of the drug. As dosing

    approaches the anticipated therapeutic range, blood, urine, and stool samples are

    collected to determine the absorption, distribution, metabolism, and excretion (ADME)

    properties of the drug and as well as the compounds into which the original drug is

    transformed while in the body. An assessment of the rate and extent of a drug's

    absorption pattern, called a bioavailability study, is also conducted. This type of study

    forms the basis for establishing bioequivalence between two brands of the same generic

    drug and can be used in an Abbreviated New Drug Application (ANDA)

    demonstrating the bioequivalence of a generic brand of a drug in comparison to a brandname of the same agent. ANDAs are often approved by FDA on the basis of

    bioavailability studies alone, saving generic drug makers the costly, time-consuming

    safety and efficacy studies required from makers of novel drug therapies.

    Following the establishment of a well-tolerated dosing regimen in Phase I studies, an

    investigational drug enters Phase II clinical trials. In these studies, the drug candidate is

    given to patients who have the condition for which the drug is intended. Phase II tests

    40

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    41/147

    are designed to determine if the tolerated dose range is therapeutically effective and if

    any adverse effects occur at an acceptable level of severity and frequency. Initially, the

    side effect frequency is determined by comparing the experimental drug to a placebo.

    Subsequently, the drug is compared to other active drugs with similar side effect

    profiles that may be metabolized in a related manner. The studies, which range from

    two weeks to three months, usually include from 200 to 300 volunteers. Shorter term

    studies are referred to as early Phase II or Phase IIA studies, while longer studies are

    designated late Phase II or Phase IIB studies. Adverse drug effects are analyzed by

    organ system (cardiovascular, respiratory, renal, skin, etc.) then assigned a severity

    rating (mild, moderate, severe) and the relationship between the reaction and the drug

    is assessed as (possible, probable, remote, or definite). In addition, the incidence of

    occurrence of the adverse effects is calculated and compared to similar data for patients

    who received a placebo. If effectiveness of the drug appears encouraging in relation to

    its side effect profile, the drug may be advanced to Phase III clinical investigations.

    Phase III clinical studies are similar to Phase II studies, but they generally continue for

    a considerably longer period of time so that the full effects of a drug candidate may be

    studied and patients long term reactions to the drug examined. This may range from

    several months to more than three years. Data on adverse reactions is typically

    evaluated for frequency, severity and relationship to the test drug.

    The New Drug Application (NDA) contains all of the drugs pertinent safety and

    efficacy data that was accumulated during its pre-clinical and clinical testing. It may

    consist of hundreds of volumes of data and is submitted to the FDA as evidence of

    safety and efficacy of the drug candidate. FDA reviews the submission and either

    grants approval of the application, states the application is approvable with the

    clarification of certain issues, or tells the NDA sponsor why it has determined that the

    NDA is not approvable.

    41

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    42/147

    Pressure to approve drug candidates

    Although FDAs drug approval process has come under heavy scrutiny since the

    market withdrawal of Mercks Vioxx in 2004, allegations of undue pressure onreviewers to approve new medicines date to well before this time.

    In 1998, for example, consumer activist group Public Citizen surveyed FDA medical

    officers who were the primary reviewers for new drug applications. Of 53 responses,

    27 cited instances in which the medical officer thought a drug too dangerous to be

    approved but approval occurred over their objection. Seventeen medical officers

    categorized the standards of FDA review for safety and efficacy as "lower" or "muchlower" compared to those in existence prior to 1995, and several stated that they had

    been instructed by their superiors to censor their reports.

    FDA found similar results in its own internal study conducted three years later. In

    2001, as FDA turnover rates among scientists and physicians rose, the agency

    conducted an assessment of job satisfaction. It found that about one-third of medical

    officers did not feel comfortable expressing differing scientific opinions, and a similar

    number felt that decisions adverse to a drug were stigmatized within the agency.

    Several reviewers said that decisions should be based more on science and less on

    corporate interests.

    A follow-up study in 2003 by the Health and Human Services Inspector General

    indicated that 18% of surveyed FDA physicians and scientists felt pressure to

    recommend that drugs be approved for sale despite their reservations about the drugs

    safety, efficacy or quality. The report concluded: "Overall, these findings present a

    significant warning signal." News reports since the Vioxx recall in September 2004

    have cited rising numbers of FDA drug safety officers claiming to have been pressured

    to approve products. A study released in July 2006 by the Union of Concerned

    Scientists found that about 15% of almost 6,000 FDA staffers polled have been "asked

    to inappropriately exclude or alter technical information or conclusions in an FDA

    scientific document." In addition, 39% of the 997 respondents stated that the FDA

    42

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    43/147

    wasn't "acting effectively to protect public health." Some FDA staff claim to have been

    punished or ignored after voicing concerns about popular medicines. For example,

    Rosemary Johann-Liang, former deputy director of the Division of Drug Risk

    Evaluation in the Office of Surveillance and Epidemiology at FDA, lost authority to

    approve strong safety recommendations after she agreed with a February 2006 review

    that recommended a black box warning for GlaxoSmithKlines diabetes medication,

    Avandia.

    Future directions in the FDA drug approval process

    The FDA has taken these cumulative allegations seriously, responding with a renewed

    effort to reduce pressure on drug reviewers and foster a culture of integrity from the top

    down. In addition to the practical goals of improving the dissemination of drug safety

    information to the public, FDAs Fall 2007 Strategic Plan specifically calls for the

    cultivation of a culture that promotes transparency, effective teamwork, and mutual

    respect, and ensures integrity and accountability in regulatory decision making. This

    encompasses the application of methods of scientific analysis with consistency,

    uniformity, and integrity with decision processes that are transparent and open to

    scrutiny, allowing for diverse points of view and vigorous debate.

    To achieve these goals, the Strategic Plan states that FDA will undertake the following

    specific actions throughout 2008:

    Develop FDA Core Values Statement: FDA Workplace Culture Initiative Co-

    Chairs will develop an FDA values statement, based on survey and focus group

    input of staff throughout the agency, to serve as guiding principles for agency

    operations. New FDA culture initiatives and the draft Values statement was

    presented at an All-Hands Broadcast by the FDA Commissioner in September

    2007.

    Develop FDA Teamwork Best Practices: The Office of Commissioner will

    coordinate a process to identify, document and share best practices that have been

    43

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    44/147

    developed by centers and offices across the agency. This will result in a reference

    list that will be compiled and shared with center and office managers.

    Strengthen FDA Advisory Committees: To ensure that the public has confidence in

    the integrity of advisory committee recommendations, the FDA has taken a variety

    of steps to increase the information about advisory committees that is made

    available to the public. It will also finalize guidance on advisory committee

    conflicts of interest.

    Given the severity of the drug safety issues that have been raised over the past several

    years, these initial steps are likely to be followed with further actions to increase the

    transparency of drug safety information and further reduce internal pressures to

    approve new medicines.

    Ensuring drug safety

    Drug safety is the primary mission of the FDAs Center for Drug Evaluation and

    Research (CDER). This group has evolved over time to address increasingly

    complicated technological, scientific and marketplace issues.

    Evolution of drug safety testing

    Drug safety testing has been an ongoing process, with a large number of laws enacted

    to strengthen FDAs oversight powers. Key developments are presented in Table 2.4.

    44

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    45/147

    Table 2.4: Timeline of key FDA legislations

    Event Impact

    Food & Drugs Act of 1906 Required drug purity

    Formation of the Food, Drug, Early framework of FDA establishedand Insecticide Administration (1927)

    Re-naming of the Food and Drug FDA formally renamedAdministration (1931)

    Federal Food, Drugs & Cosmetics Act of 1938 Required safety and efficacy testing of allmarketed drugs

    Kefauver-Harris Amendment of 1962 Required extensive animal pharmacological

    and toxicological testing before a drug couldbe tested in humans

    Federal Advisory Committee Act of 1972 Prescribed the formal use of advisorycommittees throughout the federal #government including the FDA

    Food and Drug Administration Modernization Gave manufacturers a period of marketAct (FDAMA) of 1997 exclusivity on drugs that they agreed to testin

    children

    Source: FDA Business Insights Ltd

    Although a variety of laws were in place to protect consumers prior to 1906, these did

    not specifically address drugs or medicinal products. Therefore, the environment prior

    to the turn of the century was largely unregulated and characterized by many types of

    products making unsubstantiated claims. This slowly began to change with the 1906

    passage of the Food & Drugs Act, the 1927 formation of the Food, Drug, and

    Insecticide Administration (renamed the Food and Drug Administration in 1931) andthe 1938 passage of the Federal Food, Drugs & Cosmetics Act. This latter law, which

    prohibited the manufacture and interstate shipment of adulterated and misbranded

    foods and drugs, represented a major step forward and laid the foundation for the

    U.S.s present system of drug regulation. The 1906 Act, which addressed only the

    purity of drugs, did not require that either safety or efficacy be established prior to

    marketing. This was adequate during the period from 1906 through the mid 1930s,

    since few new drugs were developed and marketed.

    45

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    46/147

    The Federal Food, Drugs & Cosmetics Act of 1938, however, required proof of both

    safety and efficacy. It followed the death of more than 100 people, many of them

    children, who had taken a new sulfa drug called Elixir Sulfanilamide. The solvent used

    in the untested product was a highly toxic chemical analog of antifreeze. Under the new

    FDCA, toxicity studies of new drugs were required prior to marketing, and the active

    ingredients in the approved product had to be listed on the label. Also, a New Drug

    Application (NDA) had to be submitted by the sponsor and approved by the FDA

    before a new drug could be promoted and distributed in the marketplace. The law

    prohibited false therapeutic claims for drugs, formally authorized factory inspections,

    and added injunctions to the enforcement tools that FDA could use to address products

    it believed were unsafe. It was not until the births of thousands of deformed infants

    whose mothers had taken the new sedative thalidomide, however, that the Act was

    strengthened to tighten control over prescription drugs, new drugs, and investigational

    drugs.

    In 1962, the thalidomide crisis resulted in the Kefauver-Harris Amendment which was

    passed unanimously by Congress. This required extensive animal pharmacological and

    toxicological testing before a drug could be tested in humans. The data from these

    studies had to be submitted in the form of a Notice of Claimed Investigational

    Exemption for a New Drug (IND) and approved by the FDA before human clinical

    studies can begin. The amendment also required that manufacturers submit to the FDA

    more substantial evidence of the investigational drug's efficacy, as well as safety, in the

    NDA. Post-approval, the amendment called for drug firms to send adverse reaction

    reports to FDA, and drug advertising in medical journals was required to provide

    complete information to the doctor including the risks as well as the benefits.

    An ongoing series of legislative and regulatory actions since the 1962 amendment have

    continued to expand FDAs role in the drug approval process. Among the more

    significant of these was the Federal Advisory Committee Act of 1972, in which

    Congress prescribed the formal use of advisory committees throughout the federal

    government including the FDA. The Act, which requires committees to be renewed

    every two years, has enabled the FDA to access a broad range of industry expertise.

    46

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    47/147

    Such committees are also responsible for a variety of critical functions, including the

    evaluation of new drug applications, Rx-to-OTC switches and issues pertaining to

    marketed drugs. For example, FDAs Center for Drug Evaluation and Research

    (CDER) currently contains 17 advisory committees delineated by therapeutic area

    which each make recommendations on new drug approvals within their class. Although

    it is not required, FDA typically follows advisory committee recommendations on such

    approvals. CDER also includes an advisory committee for Drug Safety and Risk

    Management as well as a committee for Pharmaceutical Science, which address issues

    pertaining to all therapeutic drug classes. Table 2.5shows CDER advisory committees

    operational as of March 2008.

    Under calls from industry to improve safety data on drugs used by children, legislation

    passed in 1997 provided drug makers with an additional six months of market

    exclusivity in exchange for conducting clinical trials on children. While this has

    provided important data on pediatric drug effects, which can differ significantly from

    drug effects in adults, critics of the law allege that it has given rise to speedy clinical

    trials largely designed to guarantee a positive finding for the drug.

    In February 2005, in response to concerns that FDA mishandled information about

    some drugs' potential safety risks, the Department of Health and Human Services

    announced the creation of the FDA Drug Safety Oversight Board. This new group is

    charged with monitoring and reporting information about potentially dangerous drug

    reactions. However, its role is advisory, and while members can make generally

    recommendations regarding safety issues, they do not have authority to influence a

    particular drug application, remove a drug from the market or make changes to a drug's

    sales practices.

    47

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    48/147

    Table 2.5: CDER Advisory Committees, 2008

    Advisory Committee Scope

    Anesthetic and Life Support Drugs Addresses a broad range of productsAnti-Infective Drugs Includes antibiotics, antifungalsAntiviral Drugs Includes AIDS drugsArthritis Drugs Includes COX-2 inhibitorsCardiovascular and Renal Drugs Includes hypertension drugsDermatologic and Ophthalmic Drugs Addresses a broad range of productsDrug Safety and Risk Management Addresses safety issuesEndocrinologic and Metabolic Drugs Includes weight-loss drugsGastrointestinal Drugs Includes proton pump inhibitorsMedical Imaging Drugs Addresses drugs used in diagnostic imaging

    Nonprescription Drugs Covers a broad range of therapeutic classesOncologic Drugs Focuses on cancer products

    Peripheral and Central Nervous System Drugs Addresses a broad range of productsPharmaceutical Science & Clinical Pharmacology Primarily addresses generic drugsPsychopharmacologic Drugs Includes antidepressantsPulmonary-Allergy Drugs Includes nonsedating antihistaminesReproductive Health Drugs Includes contraceptives

    Source: FDA Business Insights Ltd

    However, the FDA continues to come under criticism for approving unsafe drugs. In

    September 2006, a long-awaited review of the FDA by the Institute of Medicine claimsthat the agencys drug safety monitoring systems are inadequate and in need of reform,

    hampered by bad management and subject to incessant internal disputes. The report

    outlined a series of suggested changes including limiting initial drug approvals to five

    years. As of early 2008, however, there does not appear to be any significant attempt to

    implement this recommendation.

    Pediatric safety testing

    Because of their distinct physiologies, children often experience quite different effects

    of drugs than adults. Drugs that have been extensively studied and found highly safe in

    adults, for example, can behave quite differently in children even after dosages have

    been scaled back proportionately. At the same time, most clinical testing for new

    product candidates focuses on adults due to the far larger size of the adult population,

    as well as the significantly greater expense and heightened complexity of testing drugs

    48

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    49/147

    in children. This causes safety risks, since more than 75% of drugs that are prescribed

    to children have only been tested in adults.

    To address this problem, several initiatives have been undertaken including the Best

    Pharmaceuticals for Children Act (BPCA) and pediatric exclusivity incentives. The

    Best Pharmaceuticals for Children Act was originally enacted in 1997 and reauthorized

    in 2002. The legislation provides incentives for drug manufacturers to conduct clinical

    trials in children, gives pediatric medications priority status for FDA evaluation, and

    promotes the dissemination of information related to the use of drugs in children.

    Pediatric exclusivity provides up three years of additional market exclusivity for drugs

    whose manufacturers conduct pediatric safety studies. Through the end of 2007, these

    incentives have resulted in nearly 800 pediatric studies involving more than 45,000

    children and updated pediatric labeling for more than 120 drugs.

    Future directions in drug safety reviews

    Officially, the FDA has not changed its overall standards for drug approval in many

    years. On case by case basis, however, the agency appears to be requiring more data to

    verify safety. This is likely tied to the recent spate of embarrassing drug recalls, and is

    expected to continue into the foreseeable future. For example, the FDA is now

    requiring long term studies examining cardiovascular risk for all new non-steroidal

    anti-inflammatory drug (NSAID) candidates and in April 2007, issued a non-

    approvable letter for Mercks Arcoxia. The letter stated that Merck would need to

    provide additional data to support the COX-2 inhibitors benefit-to-risk profile, even

    though the drug was already available in 65 countries worldwide.

    In fact, even after granting approval, the FDA is expected to increase its requests for

    safety data. For example, the new FDA Amendments Act of 2007 permits the FDA to

    require companies to study the risks associated with drugs even after they have been

    approved for sale when new safety information may have emerged. The agency can

    then demand changes in product labeling, such as restrictions on use and distribution.

    49

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    50/147

    This ability was also explicitly included in the 2007 reauthorization of the Prescription

    Drug User Fee Act (PDUFA).

    In 2007, entire sections were added to PDUFA regarding post market studies, risk

    evaluation and risk mitigation. Among the most important is that drug sponsors

    respond to an FDA notice of a post market safety issue with a supplemental label

    change. FDA decides unilaterally when to make such notifications, and the

    proposed label change is subject to review and discussions. However, such discussions

    may last only 30 days following FDA notification, and within 15 days after conclusion

    of discussions, FDA may issue an order directing the labeling change. Should sponsors

    fail to respond appropriately to FDA post market safety or risk mitigation notifications,

    the drug may no longer be sold in the U.S. Congress also authorized development of

    post market risk identification and analysis methods that includes linkages among

    multiple sources of data, proposing goals of at least 100,000,000 patients by 2012.

    However, drugs and active ingredients produced outside the U.S. remain a safety risk

    and are likely to continue to do so through the foreseeable future. This is a significant

    concern, since foreign companies manufacture as much as 80% of all ingredients used

    by American drug makers. In November 2007, the director of healthcare for the

    Government Accountability Office testified before a Congressional subcommittee that

    the FDA can't guarantee the safety of the U.S. drug supply because its oversight of

    foreign drug manufacturers is lax. Due to poor record-keeping, the agency cant

    determine which facilities haven't been inspected. Furthermore, at the current rate of

    inspection, it would take the FDA 13 years to examine each known manufacturing site

    once.

    Reducing time to market

    One of the most significant mechanisms the FDA has introduced to reduce drug

    approval times has been the Prescription Drug User Fee Act (PDUFA). First

    50

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    51/147

    established in 1992, PDUFA was designed to decrease review times by strengthening

    the FDAs budget. Under the law, which was reauthorized in 1997, 2002 and 2007,

    drug makers pay fees for review of applications that cover about half of the FDA's

    budget, thereby resulting in more manpower to review applications. In 2007, these user

    fees amount to roughly $400m. The law was proposed as a means to supplement

    FDAs somewhat limited operating budget in its fiscal year 2006, for example, the

    FDA received just $1.5 billion, compared with $5.8 billion appropriated to the Centers

    for Disease Control (CDC) and Prevention and $28.6 billion for the National Institutes

    of Health (NIH). To put this in perspective, the agencys funding as a proportion of the

    CDCs budget has declined from 97% in 1986 to 39% in 1996 to 28% in 2006,

    according to the Coalition for a Stronger FDA. This dearth of resources continues to

    hamper the FDAs ability to achieve adequate staffing levels to meet its drug review

    objectives.

    In 2005, FDA made further internal changes designed to speed up the availability of

    innovative medical technologies through the creation of the Medical Innovative Task

    Force. This group proposed five recommendations including:

    Entering into new or expanded memoranda of understanding with federal agencies

    outside of HHS that play important roles in medical technology development;

    streamlining HHS involvement in medical technology;

    supporting standardization of e-clinical trials for drugs and medical devices;

    improving collaboration and cooperation between the FDA and the Centers for

    Medicare & Medicare Services (CMS) to speed approval and reimbursement

    coverage for new products;

    expanding scientific education and training to speed medical innovation.

    Progress has been made in some of these areas. For example, FDA and CMS have

    agreed to conduct parallel product reviews, at the request of an applicant. They will

    also work together on joint program to increase FDAs use of CMS post market

    51

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    52/147

    surveillance data. As a result of these and other initiatives, the FDA now attempts to

    review and act on at least 90% of the New Drug Applications and Biologic License

    Applications within 10 months of the date of filing and within six months for drugs

    given priority review.

    Fast track approvals

    In order to provide speedier approval for certain drugs that address unique needs, the

    FDA has established three categories within which applications will be reviewed on a

    shorter timetable: Fast Track, Accelerated Approval and Priority Review.

    The Fast Track process was designed to expedite the review of drugs to treat serious

    diseases for which there is an unmet medical need. If there are existing therapies, a fast

    track drug must show a significant advantage over available treatment, such as super

    effectiveness and/or reduced side effects or toxicity. A drug that meets these criteria is

    eligible to receive:

    More frequent meetings with FDA to discuss the drugs development plan and

    ensure collection of appropriate data needed to support drug approval;

    more frequent written correspondence from FDA about such things as the design of

    the proposed clinical trials;

    eligibility for Accelerated Approval;

    Rolling Review, in which a drug company can submit completed sections of its

    NDA for review, rather than waiting until every section of the application is

    completed before the entire application can be reviewed. NDA review usually does

    not begin until the drug company has submitted the entire application to the FDA.

    The Accelerated Approval process, which was first implemented in 1992, allows earlier

    approval of drugs that treat serious disease and fill an unmet need based upon a

    surrogate endpoint rather than a clinical outcome. This can considerably shorten the

    time required prior to receiving FDA approval, since surrogate endpoints (markers such

    52

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    53/147

    as a laboratory measurement, or a physical sign used in clinical trials as an indirect

    measurement of improvement) can be faster to measure while clinical outcomes such as

    survival or symptom improvement can take much longer to identify. The FDA bases its

    decision on whether to accept the proposed surrogate endpoint on the scientific support

    for that endpoint. For example, instead of waiting learn if a drug actually can extend

    the survival of cancer patients, the FDA might approve a drug based on evidence that

    the drug shrinks tumors because tumor shrinkage is considered a good predictor of

    increased survival.

    Priority Review, which was also established in 1992 with PDUFA, is given to drugs

    that offer major advances in treatment, or provide a treatment where no adequate

    therapy exists. The goal for completing a Priority Review is six months, compared

    with the 10 month goal of Standard Review. Priority Review status can apply both to

    drugs that are used to treat serious diseases and to drugs for less serious illnesses;

    however, the drug must have the potential to provide significant advances in treatment.

    This could include increased effectiveness, reduced adverse reactions, greater patient

    compliance or evidence of effectiveness in a new subpopulation, such as children.

    Orphan drug approvals

    As in Europe, U.S. regulators have focused some attention on easing the approval

    process for so-called orphan drugs. These are medicines that address diseases

    affecting fewer than 200,000 people in the United States or low prevalence is taken as

    prevalence of less than 5 per 10,000 in the community. The class was formally

    established in 1983 with the passage of the Orphan Drug Act. The act recognizes that

    medical research and development of drugs to treat such diseases is financially

    disadvantageous, so it rewards companies that do so with tax reductions and marketing

    exclusivity on such drugs for an extended time (seven years post-approval).

    Under the act, nearly 1,700 medicines have been designated orphan drugs as of 2007,

    and 249 have been approved for use. In contrast, the decade prior to 1983 saw fewer

    than ten such products come to market. In late 2007, the FDA and EMEA came to an

    53

  • 7/23/2019 Drug Approval Trend at the FDA and Emea

    54/147

    agreement whereby the same application could be


Top Related