nature reviews drug discovery (july 2012)

77
5/24/2018 NatureReviewsDrugDiscovery(July2012)-slidepdf.com http://slidepdf.com/reader/full/nature-reviews-drug-discovery-july-2012 1/77 New horizons in pharmaceutical regulation  Alasdair Breckenridge, Michelle Mello and Bruce M. Psaty A life cycle approach to pharmaceutical regulation, in which the benefit–risk balance of new drugs continues to be robustly assessed following market approval, is emerging in both the United States and Europe.  Alasdair Breckenridge is Chairman of the Medicines and Healthcare products Regulatory Agency, London SW1W 9SZ, UK. Michelle Mello is Professor of Law and Public Health at Harvard University, Cambridge, Massachusetts, Massachusetts 02115, USA. Bruce M. Psaty is Professor of Medicine, Epidemiology and Health Services at the University of Washington, Seattle, Washington 98101, USA. doi:10.1038/nrd3787 When a medicine is first marketed, much is known about its quality, pharmacology and efficacy in a selected, carefully screened group of patients involved in the clinical trial programme. However, these efficacy data may provide an incomplete, and perhaps even misleading, indication of the drug’s effectiveness in the population overall. Even greater gaps in knowledge exist with regard to its safety in the wider community. Adverse effects may be too rare to detect in pre-approval clini- cal trials or they may occur in populations that are not represented in these trials, such as patients also taking other medicines. These observations underscore the need for an ongoing assessment of a new drug’s effectiveness and safety in the post-marketing period. Importantly, the pre- occupation of the press, the public and even politicians with issues of drug safety is often misplaced, because of a focus on safety alone, even though what determines the  value of a drug is its benefit–risk balance 1 . Indeed, many drugs — for example, some commonly used anticancer agents or drugs for HIV infection — have substantial toxicities but are sufficiently effective that their benefit– risk profile supports their use in treating these serious diseases, often initially under some form of supervision to maximize the health benefits for patients. Here, we discuss new directions in pharmaceutical regulation in Europe and the United States that reflect a recognition of the need for monitoring and recalibrating the benefit– risk profile during the entire life cycle of a drug. The need for a life cycle approach The development of innovative drugs has become more challenging and more costly in recent decades for sev- eral reasons. Regulatory authorities have heightened their requirements for evidence of efficacy and safety (in part in response to major drug safety problems), and cash- strapped payers for health care increasingly need strong evidence that new drugs represent value for money com- pared with existing drugs. Indeed, industry may have shifted the emphasis of its innovative drug development into areas where the bar for approval is lower owing to a lack or absence of effective drugs, but the risk of failure is higher. Nevertheless, in some disease areas there has been considerable success, such as biologics for patients with rheumatoid arthritis and molecularly targeted drugs for some cancers (such as chronic myeloid leukaemia). However, the costs of failure can be huge, particularly for indications such as Alzheimer’s disease that require lengthy clinical trials and that lack validated surrogate end points. In the United States, and some European countries, an increasingly vocal patient advocacy movement has demanded earlier access to new medicines, particularly for life-threatening diseases. This movement, which had its origins in AIDS and cancer advocacy in the 1980s, has put regulators under pressure to relax their evidentiary requirements for the approval of potentially life-saving medicines. Most regulatory agencies have responded with accelerated approval tracks and compassionate use programmes but there are some commentators who still press for a broad reversal of a regulatory trend requiring a higher level of evidence for drug approval. For example, a former commissioner of the US Food and Drug Administration (FDA) recently called for the US Congress to allow the approval of new drugs based solely on drug safety, with efficacy to be proven in post- approval trials 2 . The US Congress is currently consider- ing legislation to speed market access of drugs for serious diseases by allowing the expanded use of surrogate end points and rapidly measurable clinical end points and thus, presumably, drug approval based on fewer, smaller and shorter trials. Such an approach, if enacted, means that regulators must move their horizons too, focusing new attention on the post-approval period of drug development with active surveillance and post-marketing clinical trials. If standards for initial drug approval are eased — and even if they are not — regulatory agencies must have a robust process in place for the continuous evalua- tion of a drug’s benefit–risk balance over its entire life COMMENT NATURE REVIEWS | DRUG DISCOVERY VOLUME 11 | JULY 2012 | 501 © 2012 Macmillan Publishers Limited. All rights reserved

Upload: krishna-maringanti

Post on 14-Oct-2015

100 views

Category:

Documents


2 download

DESCRIPTION

Drug Discovery Review

TRANSCRIPT

  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    1/77

    New horizons in pharmaceuticalregulation

    Alasdair Breckenridge, Michelle Mello and Bruce M. Psaty

    A life cycle approach to pharmaceutical regulation, in which the benefitrisk balanceof new drugs continues to be robustly assessed following market approval, is emergingin both the United States and Europe.

    Alasdair Breckenridge is

    Chairman of the Medicinesand Healthcare products

    Regulatory Agency,

    London SW1W 9SZ, UK.

    Michelle Mello is Professor

    of Law and Public Health

    at Harvard University,

    Cambridge, Massachusetts,

    Massachusetts 02115, USA.

    Bruce M. Psaty is Professor

    of Medicine, Epidemiology

    and Health Services at the

    University of Washington,

    Seattle, Washington 98101,

    USA.

    doi:10.1038/nrd3787

    When a medicine is first marketed, much is knownabout its quality, pharmacology and efficacy in aselected, carefully screened group of patients involvedin the clinical trial programme. However, these efficacydata may provide an incomplete, and perhaps evenmisleading, indication of the drugs effectiveness in thepopulation overall. Even greater gaps in knowledge existwith regard to its safety in the wider community. Adverseeffects may be too rare to detect in pre-approval clini-cal trials or they may occur in populations that are notrepresented in these trials, such as patients also takingother medicines.

    These observations underscore the need for an

    ongoing assessment of a new drugs effectiveness andsafety in the post-marketing period. Importantly, the pre-occupation of the press, the public and even politicianswith issues of drug safety is often misplaced, because of afocus on safety alone, even though what determines the

    value of a drug is its benefitrisk balance1. Indeed, manydrugs for example, some commonly used anticanceragents or drugs for HIV infection have substantialtoxicities but are sufficiently effective that their benefitrisk profile supports their use in treating these seriousdiseases, often initially under some form of supervisionto maximize the health benefits for patients. Here, wediscuss new directions in pharmaceutical regulation inEurope and the United States that reflect a recognition

    of the need for monitoring and recalibrating the benefitrisk profile during the entire life cycle of a drug.

    The need for a life cycle approach

    The development of innovative drugs has become morechallenging and more costly in recent decades for sev-eral reasons. Regulatory authorities have heightened theirrequirements for evidence of efficacy and safety (in partin response to major drug safety problems), and cash-strapped payers for health care increasingly need strongevidence that new drugs represent value for money com-pared with existing drugs. Indeed, industry may haveshifted the emphasis of its innovative drug development

    into areas where the bar for approval is lower owing to alack or absence of effective drugs, but the risk of failureis higher. Nevertheless, in some disease areas there hasbeen considerable success, such as biologics for patientswith rheumatoid arthritis and molecularly targeted drugsfor some cancers (such as chronic myeloid leukaemia).However, the costs of failure can be huge, particularlyfor indications such as Alzheimers disease that requirelengthy clinical trials and that lack validated surrogateend points.

    In the United States, and some European countries,an increasingly vocal patient advocacy movement hasdemanded earlier access to new medicines, particularly

    for life-threatening diseases. This movement, which hadits origins in AIDS and cancer advocacy in the 1980s, hasput regulators under pressure to relax their evidentiaryrequirements for the approval of potentially life-savingmedicines. Most regulatory agencies have respondedwith accelerated approval tracks and compassionateuse programmes but there are some commentatorswho still press for a broad reversal of a regulatory trendrequiring a higher level of evidence for drug approval.For example, a former commissioner of the US Foodand Drug Administration (FDA) recently called for theUS Congress to allow the approval of new drugs basedsolely on drug safety, with efficacy to be proven in post-approval trials2. The US Congress is currently consider-

    ing legislation to speed market access of drugs for seriousdiseases by allowing the expanded use of surrogate endpoints and rapidly measurable clinical end points andthus, presumably, drug approval based on fewer, smallerand shorter trials.

    Such an approach, if enacted, means that regulatorsmust move their horizons too, focusing new attentionon the post-approval period of drug development withactive surveillance and post-marketing clinical trials.If standards for initial drug approval are eased andeven if they are not regulatory agencies must havea robust process in place for the continuous evalua-tion of a drugs benefitrisk balance over its entire life

    COMMENT

    NATURE REVIEWS |DRUG DISCOVERY VOLUME 11 |JULY 2012 |501

    2012 Macmillan Publishers Limited. All rights reserved

  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    2/77

    cycle. In 2007, an expert committee of the Institute ofMedicine (IOM) in the United States characterized theFDAs approach to benefitrisk analysis as ad hoc, infor-mal and qualitative, and recommended that the agencydevelop a more systematic evaluation as part of a lifecycle approach3.

    The need for this life cycle approach again became

    apparent as a result of the recent controversy overthe thiazolidinedione antidiabetic drug rosiglitazone(Avandia), which was granted marketing authorizationin 1999 based on its ability to improve glycaemic control,as assessed by reduced levels of blood sugar and haemo-globin A1C surrogate markers that were assumed topredict clinical benefit. However, an increased risk ofcardiovascular events associated with the use of rosigli-tazone highlighted initially in 2007 became appar-ent, emphasizing the need for more effective benefitriskanalysis for recently introduced medicines.

    Emerging issues in drug regulation

    A life cycle approach has begun to emerge in pharma-

    ceutical regulation in Europe and the United States,although much remains to be done. In 2005, the EuropeanCommission proposed that a risk management planbecome part of the required regulatory submission fornew medicines. The plan should include: first, a state-ment of what is known about the safety of the medicinesand also what is not known; second, a plan for how theunknown information should be acquired; third, whatrisk minimization steps, if any, should be followed (forexample, allowing only specialists to prescribe the drugor launching an educational programme for patients);and last, how this information should be communicated.In 2008, the Commission proposed a new set of pharma-

    covigilance regulations that will take effect in July 2012.These not only reinforce the importance of risk manage-ment plans but also give regulators the authority to requirepost-marketing studies of effectiveness as well as of safety.

    In the United States, the Food and Drug Adminis-tration Amendments Actof 2007 substantially strength-ened the FDAs regulatory authority in the post-marketingperiod, allowing it to require specific studies in certaincircumstances. The FDA was also empowered to requiredrug manufacturers to submit a risk evaluation mitigationstrategy as a condition of drug approval.

    Notwithstanding these initiatives, concerns aboutdrug safety and the conduct of post-marketing studiesremained, and were heightened by the case of rosigli-

    tazone. Consequently, a second IOM committee wasconvened in 2010 at the request of the FDA and otherfederal agencies to offer further guidance on strategiesfor studying the safety of approved medicines. The finalreport4, released in May 2012, acknowledges the stepsthat the FDA has already taken to improve drug safety

    but makes 23 recommendations to implement morefully a life cycle approach to drug regulation. One ofthe most interesting recommendations is for the devel-opment of a benefitrisk assessment management plan(BRAMP) by the sponsor of every new drug. This publicdocument, which would be updated over the life cycleof the drug, would provide an updated assessment of

    its benefitrisk profile. Over time, the BRAMP wouldinclude information about any public health ques-tions posed by the drug, a formal benefitrisk assess-ment (which is regularly reviewed) and a rationale forthe type of any post-marketing study of effectivenessand safety. The European Commissions proposals andthose encompassed in a BRAMP converge in their newattention to post-marketing drug safety and an ongoingevaluation of the drugs benefitrisk profile.

    Whether the pharmaceutical industry joins in thisconsensus and whether companies will serve as willingor reluctant partners in an invigorated post-marketingdrug regulation scheme remains unclear. Additionalregulatory requirements are never welcome in the

    short term, and the post-marketing studies that maybe required under the life cycle approach may imposesubstantial costs on manufacturers. However, industryhas learned the hard way that inadequate responses tosafety signals can have a serious detrimental impact notonly on public health but also in economic and reputa-tional terms. The long-term interests of manufacturersand regulators in well-characterized benefitrisk profilesare essentially the same.

    Summary

    Experts and regulators in Europe and the United Statesalike now recognize that a robust ongoing assessment of

    the benefitrisk balance of new medicines is essential.Regulatory requirements are converging in the directionof the life cycle approach. So too, we hope, will be therecognition that a fully realized vision for drug safetyrequires the commitment of both those who regulatemedicines and those who produce them.

    1. Eichler, H.-G. et al.Safe drugs and the cost of good intentions.

    N. Engl. J. Med. 360, 13781380 (2009).2. von Eschenbach, A. Toward a 21st century FDA.Wall Street Journal

    [online], http://online.wsj.com/article/SB10001424052702303815

    404577331673917964962.html(16 Apr 2012).

    3. Institute of Medicine. The future of drug safety: promoting and

    protecting the health of the public. (National Academies Press,

    2007).

    4. Institute of Medicine. Ethical and scientific issues in studying the

    safety of approved drugs. (National Academies Press, 2012).

    Competing interests statementThe authors declare competing financial interests: see Web version for

    details.

    Disclosure and acknowledgementsThe authors served as members of the Institute of Medicine committees

    discussed in this article. They acknowledge important intellectual contri-

    butions to the ideas expressed by other members of these committees,

    but the views here are their own.

    502 |JULY 2012 |VOLUME 11 www.nature.com/reviews/drugdisc

    C OMME N T

    2012 Macmillan Publishers Limited. All rights reserved

    http://online.wsj.com/article/SB10001424052702303815404577331673917964962.htmlhttp://online.wsj.com/article/SB10001424052702303815404577331673917964962.htmlhttp://www.nature.com/nrd/journal/v11/n7/box/nrd3787_audecl.htmlhttp://www.nature.com/nrd/journal/v11/n7/box/nrd3787_audecl.htmlhttp://online.wsj.com/article/SB10001424052702303815404577331673917964962.htmlhttp://online.wsj.com/article/SB10001424052702303815404577331673917964962.html
  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    3/77

    NEWS IN BRIEF

    Antibiotic R&D gets a dose of funding

    The Innovative Medicines Initiative (IMI) has launched a 223 million (US$280 million) New

    Drugs 4 Bad Bugs (ND4BB) programme to spur the development of antibiotics.

    The lowdown:Citing an increasing risk of antibacterial resistance and a lack of progress in the

    development of new antibiotics, the IMI has launched a call for proposals for its ND4BB programme,

    which is aimed at encouraging industry and academic activity in the antibiotic space. The IMI is

    contributing 109 million to the programme, and five pharma partners (AstraZeneca, Basilea,

    GlaxoSmithKline (GSK), Johnson & Johnson (J&J) and Sanofi) are pitching in a further 114 million

    as in-kind contributions. Funders may provide further backing to the project at later stages,

    and the IMI anticipates that the programme could use up to 600 million over the next 7 years.

    ND4BB marks the first attempt by the IMI to directly fund clinical trial work. The programme

    includes a commitment to back the development of GSKs peptide deformylase inhibitor

    GSK1322322 in a Phase III trial in community-acquired bacterial pneumonia and in a Phase

    IIb trial in acute bacterial skin and skin structure infections in part in the hopes of improving

    clinical trial design and infrastructure. The programme could also fund the development of two

    AstraZeneca drugs, and back-up GSK compounds, depending on the results of these and other

    ongoing trials.

    The programme also aims to foster collaborative exchange of data and regulatoryexperiences between companies, and to build and train networks of researchers. A fraction

    of the budget is earmarked for preclinical research, including projects that could improve

    the communitys understanding of antimicrobial resistance and bacterial cell penetration,

    efflux and permeability. Full details of the first call for proposals for ND4BB are available online

    (see theIMIwebsite), and submissions are due by 9 July 2012.

    made in a plant cell system. Regulatory approval

    decisions were pending for several candidates

    including Bristol-Myers Squibb/Pfizers

    anticoagulant apixaban, Arenas anti-obesity

    drug lorcaserin and Sanofis teriflunomide for

    multiple sclerosis as Nature ReviewsDrugDiscoverywent to press.

    Last year the FDA approved 30 new drugs,

    in line with an average of 29.5 per year since

    1993, the first full year in which the Prescription

    Drug User Fee Act (PDUFA) was in place.

    A recent 5-year outlook for the industry forecasts

    that the agency will approve, on average, 3035

    new drugs per year until 2016 (Nature Rev. Drug

    Discov.11, 435436; 2012). An FDA official hassaid that the agency is anticipating that drug

    developers will file over 20 approval applications

    for cancer drugs this year.

    SGLT2, take two?

    Interest in antidiabetic sodium-dependent

    glucose co-transporter 2 (SGLT2) inhibitors has

    picked up again.

    The lowdown: Earlier this year, the FDA rejected

    AstraZenecas and Bristol-Myers Squibbs SGLT2

    inhibitor dapagliflozin, a potential first-in-class

    drug for diabetes, probably owing in part to

    concerns that it could increase the risk of

    cancer. New Phase III data and filing activity

    around Johnson & Johnsons SGLT2 inhibitorcanagliflozin are now renewing interest in

    the class of drugs, which act by blocking renal

    glucose reabsorption and thereby increasing

    glucose secretion via urine (Nature Rev. DrugDiscov.10, 645646; 2011). J&J presentedtop-line data from five trials at the American

    Diabetes Association (ADA) annual meeting

    in Philadelphia, including one trial in which

    canagliflozin plus metformin and sulfonylurea

    beat sitagliptin plus metformin and sulfonylurea

    in terms of reducing haemoglobin A1c (HBA1c)

    levels, and another in which it beat glimepiride.

    The company did not disclose cancer incidence

    data in these trials. J&J filed the drug for USapproval at the end of May.

    Dapagliflozins future in the United States,

    meanwhile, remains unclear. AstraZeneca

    and Bristol-Myers Squibb released new Phase

    III data on dapagliflozin at the ADA meeting

    showing that the drug in combination with

    sitagliptin reduces HbA1c levels versus placebo

    plus sitagliptin, but they did not release cancer

    incidence data from this trial. The Committee

    for Medicinal Products for Human Use (CHMP)

    gave the drug the green light for use in the

    European Union in April.

    First-in-class approvals include Genentechs

    Hedgehog inhibitor vismodegib for basal cell

    carcinoma, and Vertexs ivacaftor a cystic

    fibrosis transmembrane conductance regulator

    (CFTR) potentiator that is the first cystic

    fibrosis drug to address the mechanism of thedisease rather than the symptoms. Protalixs

    taliglucerase alfa, an enzyme replacement

    therapy for Gauchers disease, meanwhile

    received the first approval for a drug that is

    FDA on track with first-halfapprovals

    The US Food and Drug Administration (FDA)

    approved 12 new drugs and biologics in thefirst half of the year.

    The lowdown: The agencys Center for Drug

    Evaluation and Research (CDER) has already

    approved several novel drugs this year (TABLE 1).

    Table 1 | New drugs approved by the FDA in the first half of 2012

    Drug name Lead company Indication

    Glucarpidase* BTG International Toxic plasma concentrations of methotrexate

    Ingenol mebutate Leo Pharma Actinic keratosis

    Axitinib Pfizer Advanced renal cell carcinoma

    Vismodegib Genentech Metastatic or locally advanced basal cell carcinoma

    Ivacaftor Vertex Cystic fibrosisTafluprost Merck Elevated intraocular pressure in patients with

    open-angle glaucoma or ocular hypertension

    Lucinactant DiscoveryLaboratories

    Prevention of respiratory distress syndrome

    Peginesatide Affymax Anaemia due to chronic kidney disease

    Florbetapir F18 Avid Agent to estimate amyloid-neuritic plaque densityas Alzheimers disease diagnostic

    Avanafil Vivus Erectile dysfunction

    Taliglucerase alfa Protalix Gauchers disease

    Pertuzumab* Genentech HER2-positive metastatic breast cancer

    *These drugs were approved as biologics license applications.

    N E W S & A N A L Y S I S

    NATURE REVIEWS |DRUG DISCOVERY VOLUME 11 |JULY 2012 |507

    2012 Macmillan Publishers Limited. All rights reserved

    http://www.imi.europa.eu/content/stage-1-4http://www.imi.europa.eu/content/stage-1-4http://www.imi.europa.eu/content/stage-1-4http://www.nature.com/nrd/journal/v11/n6/full/nrd3744.htmlhttp://www.nature.com/nrd/journal/v11/n6/full/nrd3744.htmlhttp://www.nature.com/nrd/journal/v11/n6/full/nrd3744.htmlhttp://www.nature.com/nrd/journal/v11/n6/full/nrd3744.htmlhttp://www.nature.com/nrd/journal/v11/n6/full/nrd3744.htmlhttp://www.nature.com/nrd/journal/v10/n9/full/nrd3546.htmlhttp://www.nature.com/nrd/journal/v10/n9/full/nrd3546.htmlhttp://www.nature.com/nrd/journal/v10/n9/full/nrd3546.htmlhttp://www.nature.com/nrd/journal/v10/n9/full/nrd3546.htmlhttp://www.nature.com/nrd/journal/v10/n9/full/nrd3546.htmlhttp://www.nature.com/nrd/journal/v10/n9/full/nrd3546.htmlhttp://www.nature.com/nrd/journal/v10/n9/full/nrd3546.htmlhttp://www.nature.com/nrd/journal/v11/n6/full/nrd3744.htmlhttp://www.nature.com/nrd/journal/v11/n6/full/nrd3744.htmlhttp://www.imi.europa.eu/content/stage-1-4
  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    4/77

    Epizyme and Celgene have announced a

    partnership on the discovery, development

    and commercialization of inhibitors of histone

    methyltransferases (HMTs), an emerging family

    of epigenetic drug targets, for genetically

    defined cancers. Celgene, which markets

    two of the four epigenetic drugs currently

    approved the histone deacetylase (HDAC)

    inhibitor romidepsin (Istodax) and the DNA

    methyltransferase inhibitor azacitidine

    (Vidaza) gains exclusive rights outside the

    United States for Epizymes preclinical DOT1LHMT inhibitor programme against mixed

    lineage leukaemia (MLL). The deal, which

    involves an upfront payment of US$90 million

    to Epizyme, also provides Celgene with

    options to license ex-US rights to other

    Epizyme HMT inhibitor programmes, through

    which Epizyme could earn up to $160 million

    in milestone payments.

    Aberrant epigenetic changes resulting

    in gene dysregulation, such as oncogene

    overexpression or tumour suppressor

    silencing, can cause cancer. Taiping Chen,

    Associate Professor at the MD AndersonCancer Center, Texas, USA, explains that:

    Unlike irreversible genetic mutations,

    epigenetic mutations are potentially

    reversible, which makes epigenetic therapy

    an attractive approach for cancer treatment.

    Histone-modifying enzymes, including

    HDACs and HMTs, have attracted much

    interest in the context of anticancer drugs in

    recent years and, so far, two HDAC inhibitors

    are approved by the US Food and Drug

    Administration (FDA).

    Compared to HDAC inhibitors, the

    development of HMT inhibitors is much less

    advanced, but growing evidence implicates

    several HMTs as important drug targets

    in various cancers, says Yongcheng Song,

    Assistant Professor at Baylor College of

    Medicine, Texas, USA. One example is

    DOT1L, an HMT that specifically methylateshistone 3 lysine 79 (H3K79), which is a target

    for acute leukaemias with translocations of

    the MLLgene. Several predominant fusion

    partners of onco-MLL have been shown to

    recruit DOT1L, and H3K79 hypermethylation

    was also found to be a hallmark of

    MLL-rearranged leukaemias.

    Epizymes DOT1L inhibitor EPZ004777

    has been shown to selectively inhibit the

    proliferation of MLL-rearranged leukaemia cell

    lines and prolong survival in a mouse xenograft

    model of MLL (Cancer Cell20, 5365;2011).

    However, further development is neededbefore DOT1L inhibitors can become clinically

    useful drugs, says Song, in particular to

    improve on the poor pharmacokinetic

    properties of EPZ004777, which has a very

    short plasma half-life. Combination therapy

    should also be applied to further develop such

    compounds he adds. The rationale is that

    non-cytotoxic DOT1L inhibitors, which induce

    differentiation and apoptosis of leukaemic

    stem cells (or leukaemia-initiating cells), work

    slowly on this aggressive disease, and so

    combination with a cytotoxic drug that

    quickly kills non-stem leukaemia cells, thereby

    reducing overall cancer burden, should be

    useful. Chen also notes that DOT1L is widely

    expressed in human tissues and likely to be

    involved in several biological processes, raising

    questions about the potential toxicity of

    DOT1L inhibitors.With regard to targeting HMTs in

    general, a major challenge is how to find

    selective inhibitors, given that more than

    50 human HMTs use S-adenosylmethionine

    (SAM) as a common cofactor, says Song.

    However, the development of EPZ004777 and

    several other selective DOT1L inhibitors that

    target the SAM-binding site has indicated

    that ligand-based medicinal chemistry,

    guided by structure-based design, can be a

    viable approach for the discovery of potent

    and selective HMT inhibitors, he adds. Given

    that multiple companies are now developingsuch inhibitors, Chen concludes that

    targeting HMTs, as well as other emerging

    epigenetic modifiers such as lysine

    demethylases, will be a promising new

    frontier for anticancer drug discovery.

    Alexandra Flemming

    DEAL WATCH

    EpizymeCelgene deal signals interest

    in new class of epigenetic drugs

    BIOBUSINESS BRIEFS

    N E W S & A N A L Y S I S

    508 |JULY 2012 |VOLUME 11 www.nature.com/reviews/drugdisc

    2012 Macmillan Publishers Limited. All rights reserved

    http://www.sciencedirect.com/science/article/pii/S1535610811002273http://www.sciencedirect.com/science/article/pii/S1535610811002273http://www.sciencedirect.com/science/article/pii/S1535610811002273http://www.sciencedirect.com/science/article/pii/S1535610811002273http://www.sciencedirect.com/science/article/pii/S1535610811002273http://www.sciencedirect.com/science/article/pii/S1535610811002273http://www.sciencedirect.com/science/article/pii/S1535610811002273
  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    5/77

    EDITORIAL OFFICELondon

    [email protected] Macmillan Building, 4 Crinan Street,London N1 9XW, UKTel: +44 (0)20 7843 3620;Fax: +44 (0)20 7843 3629

    To subscribe and for more detailed information visitwww.nature.com/reviews/drugdisc

    chief editor:Peter Kirkpatrickenior editor:Alexandra Flemming,Charlotte Harrison, Sarah Crunkhorn,Monica Hoyos Flightenior new editor:Asher MullardAitAnt editor:Man Tsuey Tsecopy editor:Mariam Faruqienior copy editor:Catriona Rodwell,Lucie Wootton, Isabel Woodmancopy editing MAnAger:Lewis PackwoodArt controLLer:Susanne Harrisenior Art editor:Vicky Summersby,Patrick Morgan, Kirsten Lee

    MAnAging production editor: Judith Shadwellenior production editor:Simon Fenwickproduction controLLer:Natalie Smithenior editoriAL AitAnt:Laura CornseditoriAL AitAnt:Ella Linesweb production MAnAger:Dipti ShahMArketing MAnAger:Tim Redding, Nazly De La RosapubLihing director:Peter Collins

    new [email protected] Publishing Group, 75 Varick Street, 9th floor,New York, NY 100131917, USATel: +1 212 726 9200;Fax: +1 212 696 9006

    pubLiher (biophArMA):Melanie Brazil

    cutoMer ervice: [email protected]

    Copyright 2012 Nature Publishing GroupPrinted in Wales by Cambrian Printers on acid-free paper.

    peter kirkpAtrick ALexAndrA fLeMMing chArLotte hArrion

    MAn tuey teAher MuLLArdArAh crunkhorn

    EDITORS

    Over the past 25 years, monoclonal antibody-based therapeutics

    have become established as effective medicines for several

    serious diseases. However, it is only recently that antibodies

    have also emerged as valuable tools to inform small-molecule

    drug discovery. In a Perspective article, Lawson discusses the application of

    antibodies in the validation of targets and design of screening assays, as well as

    their potential to aid the identification of modulators of traditionally intractable

    targets, particularly proteinprotein interactions. The patents of several

    biological drugs, including leading monoclonal antibodies, are due to expire soon,

    providing opportunities to develop biosimilar versions. Yet, the complexity of such

    biopharmaceuticals raises novel challenges for the development and regulatory

    evaluation of biosimilars. In their Review, Jones and colleagues discuss key issues

    in assessing the comparability of biosimilars to pioneering biologics and describe

    analytical technologies that may be used to measure characteristics such as

    post-translational modifications that regulatory authorities have identified as being

    important in such comparisons. Monoclonal antibodies have a range of therapeutic

    applications in ocular disorders, particularly those targeting angiogenic factors

    in the treatment of age-related macular degeneration, diabetic macular oedema

    and retinal vein-occlusive diseases. Focusing on retinal diseases and glaucoma,

    Zhang and colleagues provide an overview of disease pathogenesis, highlighting

    recent innovations in ophthalmic drug discovery and delivery. Finally, Pratt and

    colleagues consider the limitations of current schizophrenia therapies and suggest

    that the lack of suitable preclinical models may be a key factor hampering

    the development of novel treatments. They propose how the use of new rodent

    models, in conjunction with translationally relevant end-point assessments,

    could increase the chances of success in schizophrenia drug discovery.

    Animal models of schizophrenia p560

    Analysing biosimilars p527

    Getty/LizZador

    Getty/RussellThurston

    NATURE REVIEWS |DRUG DISCOVERY VOLUME 11 |JULY 2012 |499

    IN THIS ISSUE

    2012 Macmillan Publishers Limited. All rights reserved

  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    6/77

    Malini Guha

    Although the field of anticancer

    immunotherapy has surged inrecent years with the approvals

    of Dendreons cancer vaccinesipuleucel-T for advanced prostatecancer and Bristol-Myers Squibbs

    ipilimumab for metastatic melanoma,one class of agents that haslanguished is the Toll-like receptor

    (TLR) agonists. Once included inthe US National Cancer Instituteslist of immunotherapeutic agents

    with the highest potential to treatcancer, TLR agonists have repeatedlydisappointed as systemic drugs.

    Most recently, in May, IderaPharmaceuticals top-line Phase IIresults of the oligonucleotide-based

    TLR9 agonist IMO-2055 showedthat the drug did not improveprogression-free survival in

    second-line recurrent or metastatichead and neck cancer. Merck KGaAhad been partnered with Idera on

    this project but stepped out last

    year, and the drugs future is nowuncertain. The most high-profilefailure, meanwhile, was Pfizers 2007

    discontinuation of its oligonucleo-tide-based TLR9 agonist CPG 7909

    (also known as ProMune) after thedrug failed to demonstrate efficacywhen used in combination with

    chemotherapy in Phase III trials innon-small-cell lung cancer (NSCLC).The fact that TLRs are expressed by

    some cancers, and that TLR agonistshave been found to sometimesdrive tumourigenesis in preclinical

    models although this has not beenobserved directly in the clinic hasnt helped matters (Nature Rev.

    Cancer9, 5763; 2009).Yet some experts believe that

    the many failures should not be

    attributed to the agents themselves.They werent used in the right way,says Nina Bhardwaj of the NYU

    Langone Medical Center in NewYork, USA. Although investigatorsagree that TLR agonists are unlikely

    to prove themselves in most cancers

    as monotherapies, they argue thatthe agents still offer promise if theyare better combined with anticancer

    drugs that kill cancer cells andthereby release tumour antigens

    that stimulate an immune response,or if they are used as therapeuticcancer vaccine adjuvants. The future

    is a multipronged approach, saysBhardwaj.

    Both the vaccine and

    chemotherapy approaches will havean important role, and will depend onthe specific clinical context, says Rob

    Hershberg, chief executive of VentiRx.Yet opinions diverge on whichcancer antigens, chemotherapies and

    targeted drugs to test the agonistswith. Because members of the TLRfamily each trigger the release of a

    different spectrum of inflammatorycytokines, there is also uncertaintyover which targets to agonize to evoke

    the most potent antitumour immuneresponses. Other outstandingquestions include whether the

    agonists need to be combined with

    Anticancer TLR agonists on the ropesToll-like receptor agonists have hit another setback with the Phase II failure of Ideras IMO-2055,but these immunotherapies may still make a comeback if appropriate combinations with vaccineantigens or anticancer drugs can be identified.

    NEWS & ANALYSIS

    NATURE REVIEWS |DRUG DISCOVERY VOLUME 11 |JULY 2012 |503

    2012 Macmillan Publishers Limited. All rights reserved

    http://www.nature.com/nrc/journal/v9/n1/full/nrc2541.htmlhttp://www.nature.com/nrc/journal/v9/n1/full/nrc2541.htmlhttp://www.nature.com/nrc/journal/v9/n1/full/nrc2541.htmlhttp://www.nature.com/nrc/journal/v9/n1/full/nrc2541.htmlhttp://www.nature.com/nrc/journal/v9/n1/full/nrc2541.htmlhttp://www.nature.com/nrc/journal/v9/n1/full/nrc2541.htmlhttp://www.nature.com/nrc/journal/v9/n1/full/nrc2541.html
  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    7/77

    agents that counter the immunosuppressive

    tactics of cancer and whether the fast-growingmetastatic tumours that most novel anticancerdrugs are initially investigated in are an

    appropriate testing ground. There are manyunresolved questions; we have a lot of work todo, says Bhardwaj.

    Vaccine or drug combo therapy?

    In the 1890s, surgical oncologist William

    Coley noticed spontaneous tumourregressions in some cancer patients who hadcontracted bacterial infections, and speculated

    that bacteria somehow activate antitumourimmune responses. TLRs which triggeran immune response upon detection

    of pathogens were later linked to theantitumour effect of Coleys toxin, and thebacille CalmetteGurin (BCG) tuberculosis

    vaccine, which contains a bacterium thatactivates multiple TLRs, became the first

    therapeutic to take advantage of this activitywhen used for localized bladder cancer.

    Yet although there have been a couple ofsuccess stories in localized cancer BCG and

    the topical TLR7 agonist imiquimod for basalcell skin tumours the class has failed in thetreatment of metastatic disease.

    In most of the failures, the TLR agonistswere being tested in combination withchemotherapy or targeted anticancer agents.

    Some experts consequently argue that TLRagonists are better suited as therapeutic cancer

    vaccine adjuvants, in which the agonists are

    used to activate the immune systems dendriticcells (DCs) so that they are better primed torecognize the specific tumour antigens that

    make up the core of the vaccines.Indeed, the only experimental TLR agonist

    currently in Phase III trials is being usedin a vaccine. GlaxoSmithKlines melanoma-associated antigen 3 (MAGEA3)-specific

    cancer immunotherapy (astuprotimut-R)combines a MAGEA3 antigen with anadjuvant system that includes the TLR9

    agonist VaxImmune, which is the same TLR9agonist as Pfizers unsuccessful CPG 7909, andthe TLR4 agonist MPL, which is also used

    in GlaxoSmithKlines approved Cervarixvaccine. Two large Phase III trials are testingthe vaccine in resected NSCLC and melanoma

    in patients whose tumours express MAGEA3,with the aim of reducing disease recurrence.Pivotal data are expected in 2013. MPL is also

    being used as an adjuvant in Merck KGaA/Oncothyreons Stimuvax vaccine, whichincludes the MUC-1 antigen and is in a

    Phase III trials in unresectable NSCLC.Proponents of the vaccine approach

    note that by using a single antigen, such as

    MAGEA3, they can develop therapies with a

    high specificity for tumour cells. Critics point

    to the risk that resistance may arise to a singleantigen vaccine and add that not all patientswith a given cancer usually express the same

    antigens (about one-third of NSCLC patientsand two-thirds of melanoma patients havetumours that express MAGEA3, for example).

    Chemotherapies and targeted agents,meanwhile, may result in a greater release ofdifferent cancer antigens when they kill cells,

    leading others to argue that these should beused to prime DCs that have been activated byTLR agonists. The fact that this combination

    approach has not worked so far has notdeterred a few groups from carrying on.They cite other reasons for the failures,

    including inappropriate use of chemotherapiesthat were too toxic in combination with theTLR agonist or too immunosuppressive,

    or doses of the TLR agonist that were too low.Mologens chief executive Matthias Schroff

    says, for example, that its DNA-based TLR9agonist MGN1703 can be given at a dose tentimes higher than the dose of CPG 7909 usedin Pfizers NSCLC trials. In May, Mologen

    reported potentially promising top-line resultsof a randomized 55-patient Phase II trial ofMGN1703 in metastatic colorectal cancer.

    Bhardwaj also argues that whereas TLRagonists have been delivered subcutaneouslywhen used in combination with standard

    anticancer drugs, they would fare better ifdelivered locally. Although local delivery canbe difficult or impossible, systemic delivery

    is like trying to stimulate a general immuneresponse throughout the body, hoping thatsomething will happen locally, she says.

    Others testing systemic administration say thattheir strategy should work, however, because

    activated immune cells circulate throughoutthe bloodstream and will eventually reach thetumour site.

    Another possible reason for past failures,says Hershberg, has been the focus on TLR9and TLR7 agonists. TLR9 agonists were

    pursued initially because they demonstratedantitumour activity in various preclinicalmodels, whereas TLR7 agonists were pursued

    because of imiquimods success (the drugwas found to act through TLR7 after itsclinical efficacy was demonstrated). But, he

    says, [TLR9 and TLR7] are not as potent atdriving a T cell response as TLR8 agonists,pointing to cell culture data showing that

    VentiRxs small-molecule TLR8 agonistVTX-2337 induces a stronger activation ofthe cytokine interleukin-12 (IL-12) than do

    imiquimod and CPG 7909. A 200-patientrandomized Phase II study of VTX-2337plus doxorubicin chemotherapy in metastatic

    ovarian cancer is planned for later this year.

    Cleveland BioLabs is similarly steering

    clear of the pack. Its candidate CBLB502,a recombinant derivative of the bacterialprotein flagellin, acts on TLR5 and is in

    Phase I development for solid tumours.The same question of which TLR to

    target also plagues the therapeutic vaccine

    field. Although it is difficult to compare thedifferent TLR agonists in randomized clinicaltrials, the Cancer Vaccine Collaborative

    a joint programme of the Cancer ResearchInstitute and the Ludwig Institute for CancerResearch has conducted a series of

    parallel, early-stage trials evaluating TLR3,TLR4, TLR7 and TLR9 agonists as vaccineadjuvants in combination with the cancer

    testis antigen NY-ESO-1. It found that theTLR3 agonist poly-ICLC elicited the mostfavourable immune response, as assessed by

    antigen-specific antibody levels as well as CD4and CD8 T cell responses. It is now planning

    to incorporate the agent into 1015 moreacademic clinical trials.

    Tackling immunosuppression

    Ruslan Medzhitov, from the Yale School ofMedicine in New Haven, Connecticut, USA,provides a further possible explanation for

    the disappointment of TLR agonists: theTLR system evolved primarily as a defenceagainst outside invaders rather than against

    cancers, and cancers take advantage of theconserved mechanisms that normally preventautoimmune responses to self-antigens.

    We need to figure out how to get around thislimitation with immunotherapy, he says.The immune system is not wired to fight

    against cancer, as it is in the case of infection.He and many others believe, therefore,

    that drug developers need to tackle tumourimmunosuppression when using TLRagonists. Not only does the tumour secrete

    immunosuppressive factors and activateregulatory immune system cells, but TLRagonists themselves seem to induce a negative

    feedback mechanism that probably limitsthe uncontrolled inflammation that couldotherwise arise.

    Several groups are experimenting alongthese lines. Hailing Lu, from the Universityof Washington, Seattle, USA, showed in

    a preclinical study that treatment withtopical imiquimod induced high levels ofthe anti-inflammatory cytokine IL-10, and

    blockade of IL-10 enhanced the antitumoureffect of imiquimod (J. Immunol.184,53605367; 2010). She is now hoping to use the

    same combination in a Phase I trial in breastcancer patients with chest wall metastases.Similarly, TriMod Therapeutics plans to

    initiate a clinical trial of a TLR agonist and a

    N E W S & A N A L Y S I S

    504 |JULY 2012 |VOLUME 11 www.nature.com/reviews/drugdisc

    2012 Macmillan Publishers Limited. All rights reserved

    http://www.jimmunol.org/content/184/9/5360.abstracthttp://www.jimmunol.org/content/184/9/5360.abstracthttp://www.jimmunol.org/content/184/9/5360.abstracthttp://www.jimmunol.org/content/184/9/5360.abstracthttp://www.jimmunol.org/content/184/9/5360.abstracthttp://www.jimmunol.org/content/184/9/5360.abstracthttp://www.jimmunol.org/content/184/9/5360.abstract
  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    8/77

    phosphoinositide 3-kinase (PI3K) inhibitor

    after preclinical studies showed that thiscombination suppressed anti-inflammatoryIL-10 and regulatory transforming growth

    factor (TGF) activity induced by the TLR5agonist flagellin (Cancer Res. 72, 581591;2012). Others are testing TLR agonists

    preclinically with cytotoxic T lymphocyteantigen 4 (CTLA)-targeting drugs as well.The potential danger in blocking

    immunoregulatory mechanisms, of course,is the risk of excessive immune reactions.Many people are thinking how to make these

    therapies more targeted so that we get anoverwhelming inflammatory response only atthe tumour site and not in the whole body, but

    so far we dont know how to do this, says Lu.Some in the field of cancer

    immunotherapy have also come to believe

    that rapidly progressing or bulky metastatic

    disease may not be the best setting totest immunotherapies, as numerousimmunosuppressive mechanisms are likely

    to be in play in these tumours, and inducedimmune responses take time to becomeeffective. But trials in the adjuvant setting that

    are designed to prevent disease recurrence(as is being tested with GlaxoSmithKlinesMAGEA3 vaccine) are long, expensive and out

    of reach for many small biotech companies.Within metastatic disease, fortunately, there

    are different settings, and some of these might

    be more amenable to treatment with TLRagonists than others. Mologens MGN1703,for example, was tested as a maintenance

    therapy in patients with colorectal cancerwhose tumours were already stabilized orresponded to standard therapy.

    N E W S & A N A L Y S I S

    NATURE REVIEWS |DRUG DISCOVERY VOLUME 11 |JULY 2012 |505

    2012 Macmillan Publishers Limited. All rights reserved

    http://cancerres.aacrjournals.org/content/72/3/581.abstracthttp://cancerres.aacrjournals.org/content/72/3/581.abstracthttp://cancerres.aacrjournals.org/content/72/3/581.abstracthttp://cancerres.aacrjournals.org/content/72/3/581.abstracthttp://cancerres.aacrjournals.org/content/72/3/581.abstracthttp://cancerres.aacrjournals.org/content/72/3/581.abstract
  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    9/77

    Asher Mullard

    US funders are spending US$20 million to put

    58 abandoned therapeutics, contributed by

    8 drug developers, into the hands of academic

    drug hunters. The drug repurposing pilot

    scheme which grew out of a meeting last

    year between US National Institutes of Health

    (NIH) officials, industry and academic

    researchers (Nature Rev. Drug Discov.10,

    399400; 2011) follows closely on the heels

    of a similar venture in the United Kingdom. InDecember last year, the UK Medical Research

    Council (MRC) pitched in UK10 million ($15

    million) to provide UK researchers with the

    means to study 22 compounds that have been

    de-prioritized by AstraZeneca.

    Through the open-innovation schemes,

    drug companies will pull previously shelved

    compounds from out of their freezers, and the

    US and UK funders will pay for further clinical

    research in new indications. For the NIHs

    scheme, all compounds have been tested in

    humans and so come with extensive safety

    and pharmacokinetic data that are nottypically made public but have been

    discontinued owing to either lack of efficacy

    in their initial indication or strategic

    refocusing (the asset list is available at

    go.nature.com/5sXoVG). The compounds are

    being contributed by Abbott, AstraZeneca,

    Bristol-Myers Squibb, Lilly, GlaxoSmithKline,

    Johnson & Johnson, Pfizer and Sanofi.

    AstraZeneca is sharing some of the same

    compounds with the MRC and the NIH,

    although the MRCs experiment also includes

    a subset of 12 compounds that are currently

    only deemed appropriate for preclinical

    studies (a list of the MRCAstraZeneca assetsis available at go.nature.com/kNSZYs).

    Although the contributed compounds

    span therapeutic areas, the US programme

    in particular is heavily loaded with central

    nervous system drugs(FIG. 1).Several

    companies have independently donated

    compounds from once-promising psychiatric

    drug classes including histamine receptor

    antagonists, nicotinic acetylcholine

    receptor agonists and 5-hydroxytryptamine

    receptor modulators underscoring a

    widespread disinvestment in this space

    in recent years. And these are not just clear

    reject compounds. In the case of the

    histamine receptors, Rob Leurs, a medicinal

    chemist at the University of Amsterdam,

    the Netherlands, and expert in the class,

    comments that the contributed drugs could

    unlock the enormous clinical promise that

    new histaminergic compounds once held.

    The US asset list is mostly made up of orally

    available small molecules, but also includes

    one monoclonal antibody (AstraZenecas

    MEDI2338) and one recombinant DNAplasmid (Sanofis XRP0038).

    There are plenty of potential uses that

    come to mind for most of these that have

    at least a reasonable chance of success,

    says Bryan Roth, a pharmacologist at the

    University of North Carolina, Chapel Hill,

    USA.

    Under the terms of both crowd-sourcing

    programmes, pharma companies will have an

    early opportunity to vet incoming ideas that will

    be proposed by academics and decide which

    theyd like to collaborate on, but the funders will

    independently and ultimately decide whichprojects to back. The NIH, via its new National

    Center for Advancing Translational Sciences

    (NCATS), hopes to fund around eight projects

    that have the potential to provide Phase I/II

    proof-of-principle data, although it could

    support more if individual centres at the NIH

    also decide to pitch in. The MRC, which has

    already whittled a list of over 100 initial

    proposals down to a shortlist of 25 since

    announcing their programme last year, plans to

    pay for 1015 preclinical and clinical projects.

    Responses to the programmes span the

    spectrum. I absolutely applaud this pot

    of funding, says Aris Persidis, CEO ofrepurposing biotech Biovista. What is good

    for repositioning in one arena is good for

    everybody in the field, he adds. We might

    as well give it a try and see, notes Roth.

    $20 million is really cheap as far as these

    things go. Others are more sceptical,

    with Pfizers former head of research and

    development (R&D) John LaMattina among

    others arguing that repurposing is already

    widely done in-house throughout industry,

    and questioning the focus on applied over

    basic research (go.nature.com/4TOWv8).

    Thinking big

    Funders provide several defences to their

    critics. For one, argues Kathy Hudson, acting

    deputy director of the NCATS, the NIH project

    is about more than delivering new drugs to

    patients. First and foremost we are pilot

    testing a model for multistakeholder

    collaboration, she says. Second, we are

    hoping that fantastic science will come

    out about disease mechanisms.

    Chris Watkins, head of translational

    research at the MRC, sees the UK project

    similarly. Our primary intention is not to

    repurpose these molecules; it is to use them to

    increase our understanding of mechanisms

    of human disease and novel therapeutic

    opportunities, he says. As a result, he adds,

    individual projects might validate a target for

    a specific disease and are therefore more akin

    to fundamental discovery science than to

    applied research. You can do extremely

    innovative clinical research that is discoveryscience, he says.

    When confronted with more pointed

    questions about why academia is likely to

    succeed in its attempts to reposition drugs

    after pharma scientists have failed, both point

    to the programmes unique ability to tap

    into the expertise and knowledge base of

    thousands of researchers from across

    disciplines, therapeutic areas and biases.

    So does Don Frail, a vice president at

    AstraZeneca and architect of several

    repurposing projects. This concept of crowd

    sourcing has worked really well in our earlyexperience with the MRC, he says, adding

    that all but one of the 22 MRCAstraZeneca

    assets received at least one proposal.

    Given the broad goals and early days of the

    schemes, its perhaps no surprise that even

    some supporters have quibbles about the size,

    scope and details of the programmes.

    For one, the schemes are focused on on-target

    repurposing, whereby drugs are retested on

    the basis of the activity they were designed

    to possess rather than on the basis of

    unanticipated activity. When researchers can

    make solid arguments that a specific off-target

    activity is likely for example, because of highstructural similarity between the primary target

    and a protein of interest they can apply for

    funding. But, laments Roth, investigators

    who want to hunt further afield for activity

    with exploratory target-based, phenotypic

    or computational screens are out of luck.

    It would have been great to be able to screen

    these compounds, says Roth.

    Frail defends the screening exclusions

    of both programmes by arguing that the

    discoveries of therapeutically relevant

    off-target activities are low-probability

    Drug repurposingprogrammes get lift offWhat will schemes designed to get industrys discarded drug candidates into the

    hands of academic researchers deliver?

    N E W S & A N A L Y S I S

    NATURE REVIEWS |DRUG DISCOVERY VOLUME 11 |JULY 2012 |1

    2012 Macmillan Publishers Limited. All rights reserved

    http://www.nature.com/nrd/journal/v10/n6/full/nrd3473.htmlhttp://www.nature.com/nrd/journal/v10/n6/full/nrd3473.htmlhttp://www.nature.com/nrd/journal/v10/n6/full/nrd3473.htmlhttp://www.nature.com/nrd/journal/v10/n6/full/nrd3473.htmlhttp://www.nature.com/nrd/journal/v10/n6/full/nrd3473.htmlhttp://go.nature.com/5sXoVGhttp://go.nature.com/kNSZYshttp://go.nature.com/4TOWv8http://go.nature.com/4TOWv8http://go.nature.com/4TOWv8http://go.nature.com/kNSZYshttp://go.nature.com/5sXoVGhttp://www.nature.com/nrd/journal/v10/n6/full/nrd3473.htmlhttp://www.nature.com/nrd/journal/v10/n6/full/nrd3473.html
  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    10/77

    events in his experience, especially in

    the current era of highly optimized drug

    candidates. But commercial concerns may

    also be at play. Exploration of off-target

    pharmacology is more likely to require the

    development of new drug doses, formulations

    and routes of administration, points out

    Persidis, and this can lead to the creation

    of new intellectual property (IP) that can

    undermine the originators control of a

    project. It is essentially impossible to develop

    a drug with such extreme specificity that it

    will not have some kind of off-target activity,

    and the network-pharmacology that renders

    each drug unique and also gives all of us

    opportunities, he adds.

    Another possible sticking point is whether

    patent arrangements will get in the way of

    delivering new drugs. Part of the need for the

    NIH programme, says Hudson, is that it can

    take longer to negotiate a deal between

    individual investigators and a drug company

    than it would take to do the research that

    drives the deal. Getting all parties to agree

    upfront to boilerplate agreements which

    give new use IP to the researchers while

    keeping composition of matter IP and a first

    chance to license new IP in the hands of the

    respective contributing drug firms certainly

    saves time. But the chance that successful

    findings will eventually make it to patients,

    says Persidis, may now hinge on the reversionrights that let the investigators license their

    new IP to third parties, and on whether

    contributors may block further development

    plans if they foresee commercial complications

    on the horizon.

    Hudson notes that investigators will have

    reversion rights, but the speed with which they

    will be able to take their new use IP to other

    parties remains unclear. Potential licensees

    would probably also need to license

    the composition of matter IP from the

    originators, she adds.

    Given the historic success rate of drugdiscovery, adds Hudson, patent issues may be

    the least of anyones concerns in terms of

    successfully steering the retested compounds

    towards approval. We have leapfrogged over

    6 or 7 years of preclinical and early-stage

    research and $30 million or so of investment

    with these compounds thats where the time

    saving is. But moving from here to the next

    stage will have the same failure rate as

    anybody else, she says. With the probability

    that a compound will make it to market from

    Phase I and Phase II at around 5% and 11%,

    respectively (Nature Rev. Drug Discov.10,

    328329; 2011), a scenario in which even one

    of the rescued compounds will ultimately end

    up in a medicine cabinet is not guaranteed.

    From pilot to permanent?

    A further counterargument to criticisms big and

    small is that both the NIH and the MRC see their

    ventures as pilot programmes. The scope of

    the solicited projects, the number of candidates

    made accessible, the language in the

    pre-negotiated template agreements and more

    can be reassessed once the pilots wrap up, and

    amended if everyone is still keen to continue.

    With one eye already on such a future,

    Watkins says the MRC is already in discussions

    with a number of companies that would like

    to join in on the UK venture.

    Although obvious ways to expand the

    project are to get more companies to

    contribute a bigger batch of failed drugs andto allow compound screening for potentially

    relevant off-target activity, a more ambitious

    approach might be to get companies to

    contribute their active pipeline candidates as

    well. The belief that a particular set of failed

    compounds holds promise above and beyond

    all manner of other compounds, including

    successful compounds, simply fails to pure

    logic, says Persidis. In 2010, Frail helped Pfizer

    and Washington University to strike a deal that

    provides researchers with data and access to

    over 500 compounds from across stages of the

    pipeline which was essentially a blueprintfor the new NIH and MRC schemes

    suggesting that it may eventually be possible

    to overcome the legal, regulatory and

    commercial barriers that led the NIH and MRC

    programmes to focus initially on repurposing

    failed drugs.

    I agree that the programme has to start

    from somewhere, but an over-reliance and

    over-representation of failed compounds is

    technically limiting and is a disservice to the

    possibility of finding new cures, says Persidis.

    Figure 1 | Breakdown of the US NationalInstitute of Healths repurposing assets by

    therapeutic area. Eight pharmaceuticalcompanies contributed 58 compounds that hadpreviously been tested in 90 indications. Centralnervous system (CNS) includes psychiatricindications and neurodegenerative indications;respiratory includes chronic obstructivepulmonary disease, asthma, allergic rhinitis andcough; pain includes both CNS and other typesof pain; metabolic includes diabetes andobesity; autoimmune includes rheumatoidarthritis, multiple sclerosis and Crohns disease;other includes endometriosis, irritable bowelsyndrome, gastroesophageal reflux disease andgenitourinary indications.

    Central nervous systemCardiovascularMetabolicAutoimmune disorders

    RespiratoryPainOncologyOther

    30%

    14%

    10%

    10%

    9%

    7%

    4%

    16%

    N E W S & A N A L Y S I S

    2 |JULY 2012 |VOLUME 11 www.nature.com/reviews/drugdisc

    2012 Macmillan Publishers Limited. All rights reserved

    http://www.nature.com/nrd/journal/v10/n5/full/nrd3439.htmlhttp://www.nature.com/nrd/journal/v10/n5/full/nrd3439.htmlhttp://www.nature.com/nrd/journal/v10/n5/full/nrd3439.htmlhttp://www.nature.com/nrd/journal/v10/n5/full/nrd3439.htmlhttp://www.nature.com/nrd/journal/v10/n5/full/nrd3439.htmlhttp://www.nature.com/nrd/journal/v10/n5/full/nrd3439.htmlhttp://www.nature.com/nrd/journal/v10/n5/full/nrd3439.html
  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    11/77

    In April,the Chinese State Intellectual Property (IP) Office published

    the National IP Strategy for2012, which aimed to enhance the

    quality of IP within the country.

    But parts of the plan that called for more effective use of

    compulsory licences, combined with an announcement in May

    that outlined new measures related to compulsory licence

    applications, sparked debate as to whether China was introducing

    new laws in an effort to begin making low-cost copies of medicines

    that are still under patent protection.

    But in reality, China as well as many other countries

    already has such compulsory licensing laws in place, which allow

    governments to override patents in a national emergency or cases

    of extreme urgency.

    If you compare existing patent law with the new measureson compulsory licensing, it is apparent that the latter did not add

    any material changes, rather the measures just supplemented some

    procedural rules, says DannyFriedmann, co-founder at KongHai

    Consultancy, Shenzhen, China, and author of the IP Dragon blog.

    Compulsory licenses are compliant with international laws (the

    World Trade Organizations trade-related aspects of intellectual

    property rights; TRIPS). For example, India recently issued a

    compulsory licence for the anticancer drug sorafenib, and Brazil and

    Thailand have issued licences for the HIV drug efavirenz. Compulsory

    licensing is also in the patent legislation of high-income countries

    such as Japan, Germany, the United States and Canada.

    Chinas National IP Strategy also aims to improve the IP appraisal

    and assessment system, and to guide inventors to shift the focus from

    patent quantity to quality. But Friedmann notes that the impact of the

    plan could be limited by a lack of implementation. In general, Chinas

    IP laws have already a lot of strength on paper. The problem andchallenge of most companies that are active in China is that these

    laws are insufficiently enforced on the ground.

    Two routes to patent challengecan give different results

    There are two ways of challenging a patentin the United States: through administrativere-examination proceedings at the US Patent

    and Trademark Office (PTO), or throughlitigation in the courts. A dispute relatedto a medical device patent has highlighted

    that sometimes these two routes can yield

    different results.Prior to the current case, the patent

    owned by Baxter International was

    found to be invalid when re-examined at thePTO but valid when assessed in concurrentproceedings by the US Court of Appeals for

    the Federal Circuit (CAFC).Because of this discrepancy, Baxterappealed to the CAFC, asserting that

    the PTOs decision was wrong. Perhapssurprisingly, in the current case the CAFC

    said that Baxters patent was indeed invalid seemingly going against its own previousdecision. But it said that the distinction arose

    because of differing evidence before the twoappeals; namely that the PTO used certainprior art to hold that the patent was invalid,

    yet this prior art had not been raised in courtproceedings.

    According to Jake Holdreith, an

    attorney at Robins, Kaplan, Miller & Ciresi,Minneapolis, Minnesota, USA: There isno question that ex partere-examinations

    [the type of re-examination carried out at thePTO in the current case] create headaches forinventors and patent owners who are trying

    to enforce their patents in court. For thatreason, it is a common strategy for an accusedinfringer to bring an ex partere-examination.

    However, some comfort for parties involvedin patent validity challenges will come fromthe fact that the CAFC noted that if the same

    evidence had been presented to both the courts

    and the PTO, then these two bodies would

    have arrivedat the same result.But this case might also have detrimental

    consequences for patentees. One thing the

    In reBaxter decision may do is encourageaccused infringers to draw out the courtproceeding and hang in as long as possible

    to try to get to a [validity] result from thePTO, rather than, for example, reaching asettlement, concludes Holdreith.

    In ReBaxter: http://www.cafc.uscourts.gov/images/stories/opinions-orders/11-1073.pdf

    23andMe patent creates a stir

    The announcement that consumer geneticstesting company 23andMe had been issued

    with a patent (US 8187811) related tohuman polymorphisms associated withParkinsons disease raised alarm among

    several of the companys customers that theirgenetic data was used to create the patents(see The Spittoon website).

    According to Pascal Borry, professor ofbioethics at the University of Leuven, Belgium,the use of customers personal data for

    developing patented methods is blurring theline between consumers and research subjects.There is nothing wrong [with 23andMe]

    using the direct-to-consumer genetic testingmodel for creating business revenues, but thisconflicts with the transparent, science-driven

    image 23andMe has been creating of its

    PATENT WATCH

    China aims to enhance IP laws MACMILLAN

    BRANDX

    N E W S & A N A L Y S I S

    510 |JULY 2012 |VOLUME 11 www.nature.com/reviews/drugdisc

    2012 Macmillan Publishers Limited. All rights reserved

    http://www.cafc.uscourts.gov/images/stories/opinions-orders/11-1073.pdfhttp://www.cafc.uscourts.gov/images/stories/opinions-orders/11-1073.pdfhttp://spittoon.23andme.com/news/announcing-23andmes-first-patent/http://spittoon.23andme.com/news/announcing-23andmes-first-patent/http://www.cafc.uscourts.gov/images/stories/opinions-orders/11-1073.pdfhttp://www.cafc.uscourts.gov/images/stories/opinions-orders/11-1073.pdf
  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    12/77

    research objectives. Consumers have been

    mislead in that regard, and saying that[permission] was in the informed consent[that customers signed] is not sufficient.

    The patent which claims methods ofscreening individuals for susceptibility toParkinsons disease was filed before the

    high-profile Prometheus case, which castdoubt on the patentability of diagnosticmethods (see Nature Rev. Drug Discov. 11,

    344; 2012), so it remains to be seen whetherthe 23andMe patent will survive anypotential subsequent validity challenges.

    Charlotte Harrison

    PATENT ADVISORS

    Daniel M. Becker: Dechert, Mountain View, CA, USA.Luke Kempton: Wragge & Co., London, UK.Leslie Meyer-Leon: IP Legal Strategies, Boston, MA, USA.George W. Schlich: Schlich & Co., London, UK.John A. Tessensohn: Shusaku Yamamoto, Osaka, Japan.Philip Webber: Dehns, London, UK.

    Toll-like receptor agonists andcancer

    Toll-like receptors (TLRs) which recognizeconserved microbial signature molecules have a central role in the innate immune

    response, suggesting that they could betargeted in cancer therapy. In the articleon p503 we highlight that although some

    TLR agonists have produced disappointingresults in clinical trials, these therapies couldstill hold promise if they are appropriately

    combined with anticancer drugs thatstimulate an immune response, or used asadjuvants for therapeutic vaccines. Here in

    TABLE 1we summarize international patentapplications published in the past 18 monthsrelated to TLR agonists and cancer. Data were

    researched using the SureChemdatabasefrom Macmillan publishers.

    Table 1 | Toll-like receptor agonists and cancer

    Patents Assignee Subject

    WO 2011022508WO 2011022509

    Array BioPharma Substituted benzodiazepines that act as TLR7 and/or TLR8 modulators; useful fortreating cancer, autoimmune diseases or infectious disorders

    WO 2012066336 AstraZeneca Benzylamine compounds that act as TLR7 agonists and are selective over TLR8 andHERG; useful for treating allergic diseases, autoimmune diseases, viral diseases andespecially cancer

    WO 2012066336 AstraZeneca Novel TLR7 agonists; useful for treating cancer

    WO 2012021834 Baylor Research Institute Novel vaccine adjuvants that consist of a dendritic cell-specific antibody conjugated toa TLR agonist; effective in stimulating the immune response

    WO 2011044246 Cleveland BioLabs Methods of treating cancer that involve using TLR5 in combination with a TLR agonist

    such as flagellin, which recruits cells involved in both the innate and adaptive immuneresponse to kill cancer cells

    WO 2011068233 Dainippon Sumitomo Pharma Imidazoquinoline compounds that have immunomodulating properties and act via TLR7;useful for the treatment of viral or allergic diseases and cancer

    WO 2011119759 Novartis Cysteine-based lipopeptides that act as TLR2 agonists; useful as vaccine adjuvants

    WO 2011031965 Gilead Novel modulators of TLRs; useful for treatingmelanoma, non-small-cell lungcarcinoma, hepatocellular carcinoma, basal cell carcinoma, renal cell carcinomaand myeloma

    WO 2011059505 Hemispherx Biopharma Novel double-stranded ribonucleic acids with rugged physicochemical structures thatbind and activate TLR3 with high specificity

    WO 2012027017 Idera Pharmaceuticals Oligonucleotide-based compounds that bind to and activate TLR3

    WO 2011139769

    WO 2011136828

    Johns Hopkins University Immunogenic compositions that consist of neoplastic cells expressing GM-CSF

    formulated with at least one TLR agonist; useful for treating neoplasiaWO 2011057148WO 2011049677

    Novartis Compounds and immunogenic compositions to treat disorders associated with TLR7;useful as vaccine adjuvants

    WO 2011128436 Sapienza University of Rome A combination of a HIF1 inhibitor and a TLR3 agonist; useful as an antitumour agentagainst solid tumours, particularly prostate tumours

    WO 2011139348 University of California Uses of phospholipid conjugates of synthetic TLR7 agonists (for example, in vaccines);useful for treating inflammation, cancer and pathogen infection

    WO 2012037612 University of Melbourne A method of treating or preventing cancer that uses a TLR2 agonist to stimulate aninnate immune response (but not a humoral or cellular immune response)

    WO 2012045090 VentiRx Pharmaceuticals A TLR agonist preferably a TLR8 agonist and its use in combination therapies forthe treatment of cancer

    GM-CSF, granulocytemacrophage colony-stimulating factor; HERG, ether-a-go-go-related potassium channel protein, HIF1, hypoxia-inducible factor 1; TLR,Toll-like receptor.

    Getty

    N E W S & A N A L Y S I S

    NATURE REVIEWS |DRUG DISCOVERY VOLUME 11 |JULY 2012 |511

    2012 Macmillan Publishers Limited. All rights reserved

    http://www.nature.com/nrd/journal/v11/n5/full/nrd3735.htmlhttp://www.nature.com/nrd/journal/v11/n5/full/nrd3735.htmlhttp://www.nature.com/nrd/journal/v11/n5/full/nrd3735.htmlhttp://www.nature.com/nrd/journal/v11/n5/full/nrd3735.htmlhttp://www.nature.com/nrd/journal/v11/n5/full/nrd3735.htmlhttps://surechem.com/https://surechem.com/http://www.nature.com/nrd/journal/v11/n5/full/nrd3735.htmlhttp://www.nature.com/nrd/journal/v11/n5/full/nrd3735.html
  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    13/77

    The success of some targeted anti-cancer drugs, in particular kinaseinhibitors, is known to be due to a

    certain amount of off-target activity(polypharmacology). Since kinasesignalling cascades are complex and

    interconnected, it is thought thatsimultaneous inhibition of several

    signalling nodes is needed to preventthe rapid emergence of resistance totargeted agents. However, to date,the design of anticancer agents with

    favourable polypharmacology hasbeen guided by serendipity ratherthan intent. Now, reporting in Nature,

    the groups of Shokat and Caganpresent a systems pharmacologyapproach that allows the identifica-

    tion of targets and anti-targets for therational design of drugs with an opti-mized polypharmacological profile.

    Combining aspects of target andphenotype-based drug discovery,the authors used a Drosophila melano-

    gaster model of multiple endocrineneoplasia type 2 (MEN2), kinase

    profiling and genetics in a stepwiseapproach. The MEN2 model is basedon an oncogenic RET transgene

    (RETMEN2) targeted to developingepithelia such as wing, eye and legusing thepatchedpromotor, with the

    transgene dose calibrated to permit50% survival of D. melanogasterlarvaeto pupariation and 0% survival to

    adulthood. Oral administration ofclinical kinase inhibitors such as van-detanib, sunitinib or sorafenib resulted

    in different levels of suppression ofRET-driven lethality sorafenib wasmost potent with rescue of about 17%

    of animals to adulthood. The authorsthen screened a library of compoundsthat target RET in addition to other

    classes of kinases, and they identifiedone compound, AD57, that rescued25% of the animals to adulthood.

    It suppressed defects in wing and

    eye development and hadan improved efficacyand toxicity profile

    compared to clinicalkinase inhibitors.

    In vitroexperi-

    ments showedthat AD57 also

    inhibited theviability of patient-derived cell lines withRETMEN2mutations.

    In a mouse xenograft modelof RET-driven tumours, AD57potently suppressed tumour growth

    and showed no toxicity, validatingthe D. melanogaster model as auseful tool for the discovery of

    compounds with improvedefficacy and toxicity profiles.

    Using kinase profiling

    and further genetic andpharmacological manipulationin D. melanogaster, AD57 and its

    derivatives were analysed with regardsto their kinase inhibition profiles

    and the relative contributions of theirtargets to the efficacy and toxicityof the compound. Small alterations of

    the structure of AD57 were shown tolead to considerable changes in kinaseselectivity. For example, AD58,

    a close analogue of AD57, inducedwhole-animal toxicity, providing anopportunity to explore aspects of AD

    class drug toxicity. It was found thatERK and SRC inhibition suppressedoncogenic RET signalling, whereas

    increased RAS pathway signalling con-tributed to toxicity. AD58 was found toexert its toxic effects through the inhi-

    bition of target of rapamycin (TOR),which was thought to be due to the factthat TOR signalling provides feedback

    inhibition of the RAS pathway.These insights were used to

    generate close AD57 analogues with

    optimized polypharmacological

    profiles. AD80 and AD81, in whichTOR binding was abolished, were

    shown to rescue 7090% of MEN2Drosophilalarvae, and AD80 had animproved efficacy and toxicity profile

    in mammalian MEN2 modelscompared to AD57 and vandetanib.

    The authors caution that the true

    predictive value of this approachawaits further testing. However,they point out that the development

    of drugs with optimized polyphar-macology has distinct advantagesover the related approach of using

    drug combinations, as the latter ishampered by the cost and complexityof clinical trials, complex target-

    profile interactions and differingpharmacokinetics.

    Alexandra Flemming

    ORIGINAL RESEARCH PAPER Dar, A. C. et al.Chemical genetic discovery of targets and

    anti-targets for cancer polypharmacology.

    Nature486, 8084 (2012)

    C A N C E R

    Of targets and anti-targets

    R E S E A R C H H I G H L I G H T S

    NATURE REVIEWS |DRUG DISCOVERY VOLUME 11 |JULY 2012

    Nature Reviews Drug Discovery|AOP, published online 22 June 2012; doi:10.1038/nrd3781

    2012 Macmillan Publishers Limited. All rights reserved

  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    14/77

    Previous studies have identified ade novomicrodeletion in SHANK2

    (which encodes a molecular scaf-folding protein enriched in neuronalsynapses) in autism spectrum

    disorder (ASD), but the role of thismutation in ASD pathogenesis isunclear. Now, a recent paper inNatureshows that Shank2-mutantmice recapitulate many of thebehavioural phenotypes that are

    characteristic of ASD. Moreover,the study suggests that hypofunctionof NMDA receptors (NMDARs)

    is the underlying molecular causeof impaired social interaction andshows that this impairment could

    be pharmacologically reversed.

    To elucidate the link betweenthe de novoSHANK2microdeletion

    and ASD, the authors generatedtransgenic mice that carried thesame mutation as the human

    microdeletion. These mutant mice(Shank2/mice)showed autistic-likeimpairments in social interaction

    and in learning and memory. Theyalso exhibited higher levels of anxiety,repetitive behaviours and hyper-

    activity compared to wild-type mice.At the cellular level, Shank2/

    mice had normal basal synaptic

    transmission at hippocampal SchaffercollateralCA1 pyramidal (SCCA1)synapses, and the postsynaptic mor-phology and numbers of excitatorysynapses were unchanged. However,

    synaptic plasticity, as assessed bylong-term potentiation (LTP)

    and long-term depression (LTD),was severely impaired. Furtherexperiments showed that this

    impairment was due to NMDARhypofunction and associatedimpairment of NMDAR signalling.

    To examine whether NMDARhypofunction directly contributesto ASD-like behaviours in Shank2/

    mice, the authors tested the effectof NMDAR modulators in thesemice. Intraperitoneal injection of

    -cycloserine (a partial NMDARagonist) improved social interactionof Shank2/mice. Furthermore,

    intraperitoneal injection of CDPPB

    a positive allosteric modulator ofmetabotropic glutamate receptor 5

    (mGluR5) that ultimately enhancesNMDAR function restored theimpaired LTP and LTD at SCCA1

    synapses without affecting basalsynaptic transmission, and fullynormalized NMDAR signalling.

    These CDPPB-treated Shank2/micealso had enhanced improvement insocial interaction compared with

    mice treated with -cycloserine.However, other behaviours (for

    example, anxiety-like behaviours)

    were not improved.Together, these results suggest

    a causal link between mutations inSHANK2, reduced NMDAR func-tion and impaired social behaviour.

    The authors also suggest that

    targeting mGluR5 could be a novelstrategy for treating ASD associated

    with impaired NMDAR function.

    Man Tsuey Tse

    ORIGINAL RESEARCH PAPERWon, H. et al.Autistic-like social behaviour in Shank2-mutantmice improved by restoring NMDA receptorfunction. Nature486, 261265 (2012)

    N E U R O D E V E L O P M E N T A L D I S O R D E R S

    Exploring the links betweenSHANK2 and autism

    PHOTOALTO

    R E S E A R C H H I G H L I G H T S

    NATURE REVIEWS |DRUG DISCOVERY VOLUME 11 |JULY 2012

    Nature Reviews Drug Discovery|AOP, published online 22 June 2012; doi:10.1038/nrd3780

    2012 Macmillan Publishers Limited. All rights reserved

  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    15/77

    Thyroid hormone (TH) confersseveral beneficial metabolic effects,

    including lowering of serum choles-terol and reduction of body fat, butthe therapeutic application of this

    hormone has so far been hamperedby deleterious effects on the heart,muscle and bone. There is substantial

    interest in the design of TH mimeticsthat may avoid such adverse effectsby exhibiting TH receptor isoform-

    or tissue-selectivity. Now, Grueteret al.have shown that the microRNAmiR-208a and its target MED13

    (mediator of RNA polymerase IItranscription subunit 13) function inthe heart in the control of metabolic

    homeostasis through effects onTH signalling, thus presenting new

    opportunities for harnessing thebeneficial effects of TH.

    miR-208a a heart-specific

    microRNA encoded by an intronof theMYH6gene is associatedwith cardiomyocyte hypertrophy and

    fibrosis in response to stress, and itsinhibition improves cardiac functionand survival rates in rodent models

    of heart failure. Among the strongestpredicted and validated targets ofmiR-208a is MED13, which is a com-

    ponent of the multiprotein mediatorcomplex a transcriptional co-activator that acts as a bridge between

    DNA-bound transcription factors

    and RNA polymerase. However, untilnow, the functions of MED13 in the

    heart had not been explored.The authors used pharmacologi-

    cal and genetic tools to modulate the

    expression of miR-208a and MED13in the heart. They found that micetreated systemically with a locked

    nucleic acid anti-miR-208a wereleaner than control mice and wereresistant to high fat diet (HFD)-

    induced obesity, displaying reducedfat mass. This phenotype was not due

    to a difference in food consumption

    or physical activity but was mediatedby an increase in systemic energy

    consumption. In addition, suchmiR-208a inhibition significantlyreduced serum triglyceride and

    cholesterol levels while improvingsystemic insulin sensitivity and glu-cose tolerance. Mice transgenically

    overexpressing MED13 specificallyin cardiomyocytes (Med13-TG mice)

    displayed an analogous metabolicphenotype, being similarly resistant

    to HFD-induced obesity and insulinresistance. By contrast, cardiac deletionof MED13 enhanced susceptibility of

    mice to HFD-induced obesity whileincreasing levels of triglycerides,cholesterol and glucose.

    Next, to further explore the role ofMED13 in energy homeostasis, theauthors performed microarray analy-

    sis on hearts from Med13-TG miceand controls. More than 60 genes werefound to be downregulated in the

    hearts of the Med13-TG mice, mostof which were involved in metabolismand regulated by various nuclear

    receptors. Further studies revealed

    that many of these genes were respon-sive to TH, with MED13 specifically

    acting at the TH receptor -isoform tomodulate TH-dependent responses.Importantly, there were no increases

    in heart rate in these mice, which is acommon consequence of enhancedsystemic TH signalling.

    Together, these findings reveal anovel role of the heart in systemicmetabolic control and suggest that

    miR-208a inhibition may have appli-cations in the treatment of metabolic

    disorders, without the risk of adverse

    effects associated with previous THmimetics.

    Sarah Crunkhorn

    ORIGINAL RESEARCH PAPERGrueter, C. E. et al.A cardiac microRNA governs systemic energyhomeostasis by regulation of MED13. Cell149,671683 (2012)

    FURTHER READING Baxter, J. D. & Webb, P.

    Thyroid hormone mimetics: potential applicationsin atherosclerosis, obesity and type 2 di abetes.

    Nature Rev. Drug Discov. 8, 308320 (2009)

    M E T A B O L I C D I S O R D E R S

    Heart microRNA provides newroute to metabolic control

    BRAND X

    R E S E A R C H H I G H L I G H T S

    NATURE REVIEWS |DRUG DISCOVERY VOLUME 11 |JULY 2012

    Nature Reviews Drug Discovery|AOP, published online 22 June 2012; doi:10.1038/nrd3779

    2012 Macmillan Publishers Limited. All rights reserved

  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    16/77

    Mutant tumour suppressor p53

    proteins are expressed in and regulatetumour biology in many humancancers, and so restoration of p53

    function could be a viable anticancerstrategy. Writing in Cancer Cell,Yu and colleagues used data from the

    US National Cancer institute (NCI)

    drug screen (known as the NCI60screen) to identify a compoundthat restored the structure and

    function of one of the most commonp53 mutants.

    Because the p53 status of the

    cell lines used in the NCI60 screen which determined the IC

    50(half-

    maximal inhibitory concentration)

    values of over 48,000 compoundson 60 cell lines was known, theauthors thought these data could be

    used to identify compounds thattarget tumours expressing mutantp53. They first used an in silico

    methodology that normalized theIC

    50values to statistically define

    a good response, and then scored

    compounds based on whether they

    produced a good response againstmutant p53 while not having a good

    response against wild-type p53.Three compounds with high scores

    were thiosemicarbazone compounds,

    of which two (NSC319725 andNSC319726) were shown to inhibitthe growth of mouse fibroblasts

    expressing mutant p53. NSC319726was then assayed in tumour celllines expressing different p53 muta-

    tions (in amino acid positions 175,248 or 273 of the DNA-binding

    domain). These studies showed that

    NSC319726 inhibited cellular growththe most in cells expressing the

    p53R175mutant. Further work showedthat the NSC319726-mediated reduc-tion in cell growth was mediated by

    apoptosis, which was at least partiallydependent on the presence of thep53R175mutant protein.

    Next, the authors investigatedthe mechanism of action ofNSC319726. Using conformation-

    specific antibodies, they showed

    that NSC319726 could restore the

    conformation of mutant p53R175

    protein to that of the wild-type

    protein. Furthermore, this con-

    formational change restored theDNA-binding properties of p53R175and increased levels of p53 target

    genes, indicating that NSC319726restored the transcriptional func-tions of p53R175.

    Thiosemicarbazone com-pounds are metal ion chelatorsand redox modulators. Indeed, the

    authors found that chelation ofzinc ions and modulation of cell-ular redox states had a role in the

    NSC319726-mediated inhibition ofcellular growth, and hypothesizedthat NSC319726 could act as a source

    of zinc to allow the p53R175 mutant

    to adopt a correct conformation.When the authors investigated

    the effects of NSC319726 in miceexpressing xenograft tumoursderived from human cell lines

    expressing different p53 alleles,NSC319726 (delivered byinjection) inhibited the growth

    of p53R175Hxenografts but not ofxenografts expressing other mutantp53 alleles.

    These data further highlightthat reactivation of mutant p53

    using a small molecule is a potential

    anticancer strategy. The authorsnote that, unlike previous studies,

    NSC319726 was identified froma screen that used many differentcell lines with diverse genetic back-

    grounds, which could better reflectthe tumour heterogeneity observedin the clinic.

    Charlotte Harrison

    ORIGINAL RESEARCH PAPER Yu, X. et al.Allele-specific p53 mutant reactivation. CancerCell21, 614625 (2012)

    A N T I C A N C E R D R U G S

    Reactivating p53

    PHOTODISC

    R E S E A R C H H I G H L I G H T S

    NATURE REVIEWS |DRUG DISCOVERY VOLUME 11 |JULY 2012

    2012 Macmillan Publishers Limited. All rights reserved

  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    17/77

    ERRATUM

    Busy panels recommend Pfizers tofacitinib, Arenas lorcaserin and moreAsher Mullard

    Nature Reviews Drug Discovery11, 429 (2012)

    In the story Busy panels recommend Pfizers tofacitinib, Arenas lorcaserin and more, the la st sentence was incorrect.Johnson & Johnsons rivaroxaban is under review for acute coronary syndrome, not stroke prevention in atrial fibrillation.

    This has been corrected online.

    Nature Reviews Drug Discovery|AOP, published online 1 June 2012; doi:10.1038/nrd3778

    2012 Macmillan Publishers Limited. All rights reserved

  • 5/24/2018 Nature Reviews Drug Discovery (July 2012)

    18/77

    The clinical and commercial success of biologics suchas monoclonal antibodies and recombinant versions ofendogenous proteins is transforming the pharmaceuti-cal industry. In 2010, worldwide sales of all biologicsapproached the US$100 billion mark1, and by 2015 it isexpected that more than 50% of new drug approvals willbe biologics2, rising to more than 70% by 2025 (REF. 3).As these drugs begin to come off patent, substantialopportunities exist for other companies to make copiesor generic versions of these drugs.

    For small-molecule drugs, abbreviated regulatorypathways for the development and introduction ofgeneric versions of the drug (following the expirationof patent protection on the original product) have been

    established for more than 25 years. Rather than requiringgeneric versions to undergo the same level of evaluationas the original drug, including clinical trials, abbrevi-ated approval for the same purposes is generally basedon demonstrating that the generic drug is pharmaceu-tically equivalent (that is, it contains the same activeingredient in the same purity, strength, dosage form androute of administration) and bioequivalent (that is, it isabsorbed into the body at a similar rate and extent) tothe original drug4. Consequently, abbreviated approval isconsiderably less expensive to achieve, thus dramaticallylowering the costs of generic drugs. This has led to thewidespread use of generic versions and substantial cost

    savings for health-care systems; a recent paper noted thatin 2009 almost 75% of small-molecule drug prescriptionsdispensed in the United States were for generics, and theapproval of a generic drug resulted in average savings of77% of the originalproducts cost within 1 year5.

    However, for biologics, establishing a regulatory path-way for the introduction of follow-on versions of theoriginal product (once its patent protection has expired)is much more challenging than for small molecules. Somesimple biologics for example, small peptides such asrecombinant insulin and recombinant human growthhormone can be well characterized by establishedanalytical approaches, which has facilitated the regulatoryapproval of follow-on versions under abbreviated path-

    ways (based in part on data from the original drug and inpart on analytical data and limited clinical data in somecases)4; however, many biologics such as monoclonalantibodies and other recombinant therapeutic proteinsare much larger and more complex. For such biologics,the extent to which existing analytical technologies canbe used to support the likelihood of clinical comparabilitybetween a follow-on version and the original product ismuch more limited than for small-molecule drugs, and itis not possible to demonstrate that the two products areabsolutely identical.

    Consequently, a key question for the developmentand regulation of follow-on biologics also known as

    1Analytical Development,

    Biogen Idec, 14 Cambridge

    Center, Cambridge,Massachusetts 02142, USA.2Department of Chemistry

    & Chemical Biology,

    The Barnett Institute of

    Chemical & Biological

    Analysis, Northeastern

    University, 360 Huntington

    Ave., Boston,

    Massachusetts 02115, USA.3Waters Corporation,

    34 Maple Street, Milford,

    Massachusetts 01757, USA.

    Correspondence to G.B.J.

    e-mail:[email protected]

    doi:10.1038/nrd3746

    Analytical tools for characterizingbiopharmaceuticals and theimplications for biosimilarsSteven A. Berkowitz1, John R. Engen2, Jeffrey R. Mazzeo3and Graham B. Jones2

    Abstract | Biologics such as monoclonal antibodies are much more complex thansmall-molecule drugs, which raises challenging questions for the development and regulatory

    evaluation o