throbber
PERSPECTIVES
`
`O P I N I O N
`
`Can the pharmaceutical industry
`reduce attrition rates?
`
`Ismail Kola and John Landis
`
`The pharmaceutical industry faces
`considerable challenges, both politically
`and fiscally. Politically, governments around
`the world are trying to contain costs and,
`as health care budgets constitute a very
`significant part of governmental spending,
`these costs are the subject of intense
`scrutiny. In the United States, drug costs
`are also the subject of intense political
`discourse. This article deals with the fiscal
`pressures that face the industry from the
`perspective of R&D. What impinges on
`productivity? How can we improve current
`reduced R&D productivity?
`
`The average life expectancy of humans has
`gone up from about 45 years of age at the start
`of the twentieth century to about 77 a century
`later. This is a consequence of a number of
`factors, including increased medical know-
`ledge, better technologies and surgical tech-
`niques, better health care, better public health
`and the discovery of drugs such as aspirins,
`antibiotics, the statins, and numerous other
`such innovative and crucial medicines from
`the pharmaceutical industry. However, the
`current challenges facing the pharmaceutical
`industry are unprecedented in its history.
`Perhaps most foremost among these are the
`industry’s lower revenue growth, poor stock
`performance, the lowest number of new
`chemical entities (NCE) approvals and the
`poor late-stage R&D pipelines prevalent
`throughout the industry.
`In 2002, overall top-line revenue growth
`in the pharmaceutical industry was just
`8% and improved only slightly in 2003 to
`
`approximately 9%. Similarly, in 2003 large
`pharma stock prices were among the worst
`performing sector on the New York Stock
`Exchange (NYSE), with an average apprecia-
`tion of 0.3%, compared with the general
`S&P500 market appreciation of 26%. At
`present the average price to earnings (P/E)
`ratio of large pharma stocks is trading at a
`discount to the entire market. By contrast,
`this sector has historically traded at a pre-
`mium to the rest of the market, mainly
`because of pipeline valuations.
`
`Depressing approval rates
`In 2002, the US FDA approvals of NCEs were
`lower than at any other time in the past
`decade, and a total of just 17 NCEs were
`approved; the situation improved marginally
`in 2003 to 21 approvals. Even if biologics and
`NCEs are considered together, the number of
`FDA approvals were at their lowest since
`1994. The situation is even bleaker when the
`number of innovative NCEs approved by
`regulatory authorities are factored into this
`performance. Prous Science1 reported that
`in the eleven-year period 1990–2000 inclusive,
`the year with the lowest number of NCEs
`approved with a novel mechanism of action
`was 2000. These data are further substantiated
`by the number of FDA priority reviews of
`NCEs (an indirect measure of innovativeness
`or addressing true unmet medical need), in
`which 2002 and 2003 showed lower numbers
`of such reviews than any two-year rolling
`period in the preceding ten years2.
`This lower rate of success in the past few
`years could be accounted for, in part at least,
`
`by a number of explanations: the industry is
`currently attacking diseases of great com-
`plexity; the entry bar for new drugs is higher
`because they are often competing with
`enhanced standard of care; and/or the regu-
`latory authorities are more demanding.
`Whatever the case, these features define the
`new playing field on which the industry has
`to compete to produce NCEs that are required
`to achieve necessary growth; an examination
`of the factors that impact R&D success is
`therefore crucial in terms of devising a
`strategy that can build a pipeline needed to
`sustain the business case for large pharma.
`
`Defining the business case
`A recent survey by Accenture3 defined the
`business case for large pharmaceutical com-
`panies in terms of NCEs required to remain a
`growth company on the basis of their current
`revenues and their desired percentage growth
`(TABLE 1). On the basis of this calculation,
`Pfizer, with pharmaceutical revenues in 2003
`of approximately US $45 billion, will need to
`generate approximately nine high-quality
`NCEs per annum. GlaxoSmithKline, with
`revenues in excess of £18 billion (~ US$ 32
`billion), will need to generate about six high-
`quality NCEs per annum, and Merck, with
`US $22.5 billion in revenues, will need
`approximately 4.5 NCEs. The next tier (in
`terms of revenues) would need to deliver
`between three and four NCEs per annum and
`even the smaller companies in the top ten
`would need to deliver approximately two
`NCEs per annum.
`
`Rates of attrition
`FIGURE 1 analyses success rates from first-in-
`man to registration during a ten-year period
`(1991–2000) for ten big pharma companies in
`the United States and Europe. The data
`indicate that the average success rate for all
`therapeutic areas is approximately 11%; or,
`put another way, in aggregate only one in nine
`compounds makes it through development
`and gets approved by the European and/or the
`US regulatory authorities. More interestingly,
`
`NATURE REVIEWS | DRUG DISCOVERY
`
`VOLUME 3 | AUGUST 2004 | 7 1 1
`
`NOVARTIS EXHIBIT 2064
`Breckenridge v. Novartis, IPR 2017-01592
`Page 1 of 5
`
`

`

`P E R S P E C T I V E S
`
`$45 billion
`$30 billion
`$20 billion
`$15 billion
`
`Table 1 | NCEs required to achieve specific real growth targets as a function of 2002 revenues*
`2002 sales‡
`Anticipated sales
`Annual real
`Sales gap for
`Estimated number
`from current
`growth target
`new products
`of NCEs required to fill
`products in 2012
`to fill in 2012
`gap (over ten years)
`$30 billion
`$43.5 billion
`75–90
`$20 billion
`$29 billion
`50–60
`$13.3 billion
`$19.3 billion
`33–40
`$10 billion
`$22 billion
`40–45
`$17 billion
`30–35
`$14.5 billion
`25–30
`$12 billion
`20–25
`$ 9.67 billion
`16.5–20
`
`5%
`5%
`5%
`8%
`6%
`5%
`4%
`5%
`$6.67 billion
`$10 billion
`*Adapted from REF. 3. ‡All figures in US $. NCE, New Chemical Entity.
`
`Year 2012 required
`NCE output
`
`9.5–11
`6.5–7.5
`4.3–5
`5.5–6.0
`4.0–4.5
`3.25–3.75
`2.5–3.0
`2.15–2.25
`
`59%. Approximately 62% of all compounds
`that enter Phase II trials undergo attrition,
`and again the highest rate of attrition at this
`phase is in the oncology field: more than 70%
`of oncology compounds fail in this phase. It is
`therefore crucial that the industry develop
`and embrace paradigms (such as obtaining
`proof of concept in man early in develop-
`ment) and methodologies to identify risk
`preclinically, and to couple this with experi-
`mental medicine procedures to interrogate
`such risks in man.
`
`Underlying causes of attrition
`An examination of the root causes of why
`compounds undergo attrition in the clinic is
`very instructive and helps in the identification
`of strategies and tactics to reduce these rates
`and thereby improve the efficiency of drug
`development. The data in FIG. 3 show the rea-
`son why compounds undergo attrition and
`how this has changed over time. In 1991,
`adverse pharmacokinetic and bioavailability
`results were the most significant cause of
`attrition, and accounted for ~40% of all attri-
`tion. By 2000, these factors had dramatically
`reduced as a cause of attrition in drug develop-
`ment, and contributed less than 10%. These
`data provide further compelling evidence that
`the industry can identify and remedy the
`causes of attrition. It might also, however, be
`that the solving of this problem has signifi-
`cantly shifted the temporal attrition profiles to
`later stages, because pharmacokinetic/bioavail-
`ability failures would have occurred in Phase I
`mainly and this might now result in com-
`pounds progressing to Phases II and III and
`failing there for other reasons.
`The major causes of attrition in the clinic
`in 2000 were lack of efficacy (accounting for
`approximately 30% of failures) and safety
`(toxicology and clinical safety accounting for a
`further approximately 30%). The lack of effi-
`cacy might be contributing more significantly
`
`the success rates vary considerably between the
`different therapeutic areas: cardiovascular, for
`instance, have a ~20% rate of success, whereas
`oncology and central nervous system (CNS)
`disorders have ~5% and ~8% success, respec-
`tively. Any R&D portfolio, therefore, would
`need to aggregate the percent success based on
`the weight of the various therapeutic areas to
`calculate how many first-in-man studies are
`needed to approximate the requisite business
`case for growth.
`The high rate of attrition in drug develop-
`ment and the need for efficiency, both in
`terms of real and opportunity costs, becomes
`even more compelling when one considers
`where most of the attrition occurs in the
`pipeline. In 2001, the costs of discovering and
`developing a drug were of the order of US
`$804 million4; current estimates are closer to
`about US $900 million; considerably more of
`these costs are incurred later in the pipeline,
`and the vast majority of attrition occurs in
`full clinical development (Phases IIb and III).
`
`FIGURE 2 illustrates the top 10 drug companies’
`success and failure rates from 1991 to 2000
`across different therapeutic areas.
`The failure rate of compounds even at the
`registration stage is 23%; that is, roughly one
`in four compounds fail after all the trials and
`the documentation for submission have been
`completed, thereby incurring the full dis-
`covery and development costs and the oppor-
`tunity costs, which, on average, could be as
`much as 12 years 10 months (the average time
`taken for the development of all the drugs
`that gained approval in 2002)5. In some thera-
`peutic areas, such as woman’s health, the failure
`rate is as high as 42%, and in oncology it is as
`high as 30%. Even the rate of failures in
`Phase III trials — by which stage significant
`amounts of the costs of discovering and
`developing a drug would have been incurred
`— is far too high: approximately 45% of all
`compounds that enter this phase of full devel-
`opment undergo attrition and in some thera-
`peutic areas, such as oncology, it is as high as
`
`11%
`
`20
`
`15
`
`10
`
`5
`
`Percentage of success
`
`0
`
`
`
`
`All
`
`
`
`
`
`
`
`
`
`CNS Infectious
`Cardio-
`Arthriris
`Urology Women's
`Metabolic
`Oncology Opthal-
`disease
`vascular
`and pain
`health
`disease
`mology
`Figure 1 | Success rates from first-in-man to registration. The overall clinical success rate is 11%.
`However, if the analysis is carried out by therapeutic areas, big differences emerge. The data are from the
`ten biggest drug companies during 1991–2000. (The companies are AstraZeneca, Bristol-Myers Squibb,
`Eli Lilly, F. Hoffman-LaRoche, GlaxoWellcome, Johnson & Johnson, Novartis, Pfizer, Pharmacia,
`Schering-Plough and SmithKline Beecham; data were obtained by Datamonitor in the Pharmaceutical
`Benchmarking Study). CNS, central nervous system.
`
`712 | AUGUST 2004 | VOLUME 3
`
`www.nature.com/reviews/drugdisc
`
`NOVARTIS EXHIBIT 2064
`Breckenridge v. Novartis, IPR 2017-01592
`Page 2 of 5
`
`

`

`P E R S P E C T I V E S
`
`0
`10
`20
`30
`40
`50
`60
`70
`80
`90
`100
`
`Failure rate (%)
`
`Phase I
`
`Phase II
`
`Phase III
`
`a
`
`Success rate (%)
`
`100
`90
`80
`70
`60
`50
`40
`30
`20
`10
`0
`
`
`Central nervous system
`
`Metabolic disorders
`
`Registration
`
`
`
`Infectious disease
`Urology
`
`to therapeutic areas in which animal models
`of efficacy are notoriously unpredictive6, such
`as CNS and oncology, both of which have
`relatively higher failure rates in Phase II and III
`trials. In the case of oncology, small Phase II
`trials looking at tumour regression in small
`cohorts of patients with different tumour
`types does not always translate to outcomes
`subsequently obtained in larger Phase III
`trials. Nevertheless, in general, failures due to
`lack of efficacy and safety demonstrate the
`need for the development of more predictive
`animal models where possible and, more
`importantly, the need to develop experimen-
`tal medicine paradigms that are more pre-
`dictive of outcomes and to carry out such
`proof-of-concept clinical trials much earlier
`in development.
`
`Can success be increased?
`Several strong strands of evidence indicate
`that it is possible. First is the fact that different
`therapeutic areas have different rates of success
`and this implies that if we understood the
`inherent factors that make one area successful
`as compared with another, we could then
`attack such factors.
`Second is the finding that biologicals have
`a higher rate of success from first-in-man to
`launch — approximately 24%7. It is true that
`most biologicals have been generated in the
`areas of immunology and cancer, but the aver-
`age rate of these two therapeutic areas should
`even out to ~11% (16% for arthritis and pain
`and 5% for cancer, based on the data in TABLE 1,
`which averages to ~11% if the two were in
`equal parts).
`Third, licensing-in compounds has a con-
`sistently higher probability of success in most
`studies, at approximately 24%7. This is the case
`even if the compounds are categorized by
`the stage that the licensing-out company has
`categorized them. This phenomenon cannot,
`therefore, be attributed purely to the fact that
`the licensing-in companies gather more data
`or because they usually put the compound at
`an earlier stage in the pipeline.
`Fourth, companies with R&D budgets of
`less than US $400 million also have higher
`success rates of approximately 18%7. This
`could partly be explained by the possibility
`that these smaller companies might be more
`inclined to work on me-too drugs (which
`should have a higher rate of success), and that
`their portfolios could be more skewed towards
`one therapeutic area or another with a greater
`probability of success. However, if one con-
`siders that many of the biotech companies fall
`into these categories, that many biotech com-
`panies are working in high-attrition-rate
`therapeutic areas such as cancer, and that
`
`
`Arthritis and pain management
`
`Oncology
`
`Women's health
`
`
`Cardiovascular
`
`Opthalmology
`
`All
`
`100
`90
`80
`70
`60
`50
`40
`30
`20
`10
`0
`
`b
`
`Success rate (%)
`
`I
`
`Reg.
`III
`II
`Stage of development
`
`App.
`
`many of these companies are indeed working
`on innovative mechanisms of action, then
`clearly this cannot be the whole explanation.
`The rate of attrition of compounds with
`novel mechanisms of action is higher than
`that of those with previously precedented
`mechanisms of action (a precedented mecha-
`nism of action is defined as one hitting a thera-
`peutic target that a drug in the market place
`hits, or which has shown proof of concept in
`late clinical trials).
`Last, even comparable large companies
`with extensive portfolios that would average
`out the differences in success between differ-
`ent therapeutic areas, and therefore portfolio
`success, have different probabilities of success.
`For instance, data from the 2002 Certified
`Medical Representatives Institute survey shows
`that the success rate that Merck enjoyed from
`first human dose to market was approximately
`twofold greater than the aggregate of the six
`companies in the same cohort with R&D
`budgets of >US $2 billion per annum8. On the
`other hand, in a briefing to analysts on 17 June
`2003 Pfizer’s current President of Research
`and Development, John La Mattina, was
`quoted as saying “Right now, only one in 25
`early candidates survives to become a pre-
`scribed medicine. We think we can improve
`those odds to one in ten and greatly enhance
`our ability to bring new medicines to patients
`
`Figure 2 | Success rate by phase of
`development and by therapeutic area.
`a | Data are shown as percent success or percent
`attrition (second X axes) of compounds entering
`that particular phase of development by certain
`therapeutic areas and by the total aggregate for
`that particular phase of development. The data
`clearly show that different therapeutic areas have
`greatly different success or attrition rates, and
`that significant attrition occurs late in the pipeline.
`b | Shows the percentage rate of success of
`compounds entering first in man that progress to
`subsequent development phase. App, approval;
`Reg, registration.
`
`around the world. ”; Pfizer’s India Homepage
`states that “approximately 1 out of every 15
`drug candidates entering development com-
`pletes phase III evaluation and obtains
`approval, ” both suggesting that their rate of
`attrition might be 93–96%. These five factors
`therefore provide compelling evidence that the
`rate of attrition could be significantly reduced
`and that drug development per se does not
`have this current high attrition rate as an
`inherent constraint. Indeed, it points to the
`idea that a systematic evaluation of the sci-
`ence, strategy and processes currently used
`in drug development merit rigorous evalua-
`tion, critical appraisal and modification to
`fulfil the onerous business case demanded
`by our patients, shareholders, consumers
`and governments worldwide.
`
`How can attrition be reduced?
`Several companies in the industry are now
`beginning to take on this problem and are
`starting to make progress. Below we propose
`some approaches that are likely to be valuable,
`but this is clearly not an exhaustive list. It is
`important that the mindset of reducing attri-
`tion in development should be in place from
`the earliest stages of discovery.
`For instance, building the need to get
`very strong evidence for proof of mecha-
`nism into the discovery paradigm is crucial,
`
`NATURE REVIEWS | DRUG DISCOVERY
`
`VOLUME 3 | AUGUST 2004 | 7 1 3
`
`NOVARTIS EXHIBIT 2064
`Breckenridge v. Novartis, IPR 2017-01592
`Page 3 of 5
`
`

`

`much earlier in the drug discovery process,
`and/or by better due diligence with respect
`to competitor development programmes
`and the likelihood of true differentiation
`from such drugs that might be ahead in
`development.
`
`Future perspectives
`The demands on pharmaceutical companies
`to meet their business objectives, as well as
`the demands of consumers for cost contain-
`ment of prescription medicines, is forcing
`the industry to think about ways that effi-
`ciencies can be achieved. A particular
`emphasis is being placed on R&D because of
`the relatively dry late-phase pipelines, the
`spiralling costs of drug discovery and drug
`development, and the patent expirations of
`major blockbusters innovated in the past
`two decades. These pressures inevitably lead
`to a healthy evaluation of the science, strate-
`gies and processes involved in drug develop-
`ment, because the rate of attrition in drug
`development is simply too high, which
`makes the R&D process inefficient; effi-
`ciency and sustained profitability by the
`pharmaceutical industry are important for
`reinvestment in further R&D so that thera-
`pies for debilitating human diseases can
`continue to be developed and the price of
`medications contained.
`This inefficiency becomes even more
`acute when one considers the number of
`compounds that undergo attrition in pre-
`clinical research, and that only three out of
`every ten drugs that makes it to market
`recover the original investment made in
`them. Factors that clearly affect attrition
`rates will lead to a more efficient industry
`and will benefit shareholders, and, more
`importantly, patients and the community.
`The industry will be forced to focus on
`attrition rates to balance the costs of drug
`development, to explore cost containment
`measures while still investing significantly in
`R&D, and to continue to generate share-
`holder value. Scientific and technological
`innovations that affect efficacy and safety
`(factors that most significantly contribute to
`attrition in the clinic) will have to be
`addressed. These include more appropriate
`animal models; biomarkers that can report
`the hitting of the molecular target in dose-
`ranging, efficacy and toxicity studies; and a
`new paradigm for drug development that
`will give early readouts for proof of concept
`and one that will allow attrition to occur
`much earlier.
`We believe that governments and con-
`sumers want to reward truly innovative drugs,
`and/or those that are genuinely differentiated
`
`D
`
`1991
`2000
`
`P E R S P E C T I V E S
`
`50
`
`40
`
`30
`
`20
`
`10
`
`0
`
`Attrition for each criterion (%)
`
`Clinical
`safety
`
`Efficacy
`
`Formulation
`
`PK/
`bioavailability
`
`Commercial Toxicology
`
`Cost
`of goods
`
`Unknown/
`other
`
`Figure 3 | Reasons for attrition (1991–2000). PK, pharmacokinetics.
`
`and therefore showing that modulation of a
`target in a specific or important disease path-
`way might reduce the attrition of a large
`percentage of compounds that fail because of
`lack of efficacy. The development of imatinib
`(Gleevec; Novartis), for example, was based
`on the targeting of a very specific lesion
`(the BCR-ABL chromosomal translocation
`protein-product or Philadelphia chromo-
`some) that occurs in chronic myelogenous
`leukaemia. We have, in a similar manner,
`provided very strong evidence that inhibition
`of β-secretase inhibits the production of
`amyloid-β in knockout mice9 and that cath-
`epsin K is involved in bone resorption (further
`compelling proof of mechanism is provided
`by humans with pycnodysostosis10,11). How-
`ever, we will have to await approval of thera-
`peutics aimed at these latter two mechanisms
`to see whether drug approvals are eventually
`obtained — for example, cathepsin K is in
`Phase II trials and the impact of this approach
`on attrition is still to early to fully evaluate.
`A second method of reducing attrition is to
`eliminate compounds that have mechanism-
`based toxicity; this risk can be rigorously
`interrogated during discovery using tools
`such as gene knockouts and RNA interfer-
`ence, and, crucially, during preclinical devel-
`opment in toxicity testing. Additional tools
`such as transcriptional profiling can also
`affect attrition due to toxicity by giving specific
`gene-signature readouts that are predictive
`of toxicities obtained by previous compounds
`targeting specific molecular targets that
`have failed, and/or molecular signature algo-
`rithms that have been trained from preclinical
`toxicity studies.
`Third, an important clinical tool that can be
`used is to identify biomarkers that signal cor-
`rect dosing and whether the specific molecular
`target has been hit in early proof-of-concept
`clinical trials.
`
`Fourth, and most important, is the
`design of proof-of-concept clinical trials
`during first-in-man studies. This has the
`distinct advantage of providing evidence in
`man that the molecular target is being hit
`and that hitting such a target gives the antici-
`pated physiological response. Appropriately
`designed proof-of-concept studies (or
`experimental medicine paradigms) could
`reduce attrition due to lack of efficacy
`mostly seen in later development, and also
`have the distinct advantage of allowing
`attrition to occur earlier, which is beneficial
`both in terms of real and opportunity costs.
`This is likely to be important given that lack
`of efficacy accounts for about 30% of attrition
`in this study.
`Fifth, another important tool is the use
`of appropriate animal models for efficacy
`testing in preclinical studies. It is interesting
`that oncology and CNS —two therapeutic
`areas with very high attrition rates in the
`data provided here — are also the areas in
`which animal models are not very predic-
`tive of the true human pathophysiology.
`For example, most pharmaceutical compa-
`nies still use xenograft models for oncology
`testing, in which a tumour cell line that might
`have little relevance to the tumour in vivo is
`injected into a nude mouse (which does not
`resemble the immunology of the host; nor
`does the artificial location of the tumour
`significantly resemble what happens in vivo
`during tumorigenesis). The use of appro-
`priate genetic models (for example, trans-
`genic and gene knockout animals) of
`tumorigenesis might be more pathophysio-
`logically relevant.
`Last, another area in which attrition can
`be reduced is the discontinuation of com-
`pounds for commercial reasons either by
`gaining alignment between the research,
`development and marketing functions
`
`714 | AUGUST 2004 | VOLUME 3
`
`www.nature.com/reviews/drugdisc
`
`NOVARTIS EXHIBIT 2064
`Breckenridge v. Novartis, IPR 2017-01592
`Page 4 of 5
`
`

`

`from existing drugs and that address a true
`unmet medical need; this provides a tremen-
`dous incentive for the pharma industry to
`conduct R&D in this arena, and this in itself
`could affect R&D productivity. Drugs that
`target novel mechanisms have higher attrition
`rates12, but a combination of better-validated
`preclinical targets that have significant pre-
`clinical proof of principal, and the scientific
`and technological innovations that posi-
`tively affect efficacy and safety of drugs dis-
`cussed earlier in this article, can mitigate
`such attrition risks. It is clear that in
`the twentieth century the pharmaceutical
`industry has had significant positive impact
`on the health and longevity of humans
`across the globe, but the early twenty-first
`century will demand both great effective-
`ness and efficiency from the industry, and it
`is therefore vital that the industry rapidly
`gears up to meet these demands.
`
`Ismail Kola, Ph.D. (Med), is Senior Vice-President
`of Basic Research at Merck Research Labs, 126 East
`Lincoln Avenue, Rahway, New Jersey 07075, USA.
`John Landis , Ph.D., is Senior Vice-President
`Pharmaceutical Sciences and Compliance Clinical
`Sciences at Schering-Plough Research Institute,
`2000 Galloping Hill Road, Kenilworth New Jersey
`07033, USA. Correspondence to I.K.
`e-mail: ismail_kola@merck.com
`doi:10.1038/nrd1470
`Kola, I. & Rafferty, M. New technologies that may impact
`drug discovery in the 5–10 year timeframe workshop.
`2002 Biomed. Expo Ann Arbor Michigan (data derived
`from Prous Science, Drugs News Prospect (2002).
`Franz, S. 2003 approvals: a year of innovation and upward
`trends. Nature Rev. Drug Discov. 3, 103–105 (2004).
`Accenture Consulting. High performance drug discovery:
`an operating model for a new era. Accenture (2001).
`DiMasi, J. A., Hansen, R. W. & Grabowski, H. G.
`The price of innovation: new estimates of drug
`development costs. J. Health Econ. 22, 151–185 (2003).
`Fearn, C. J. IMS Health. The World Pharmaceutical Market
`Presented at the Strategic Management Review, Australia,
`19–20 June (2002).
`Booth, B., Glassman, R. & Ma, P. Oncology’s trials.
`Nature Rev. Drug Discov. 2, 609–610 (2003).
`Benjamin, G. A. & Lumley, C. E. Industry Success Rates
`2003 Including Trends in Success Rates CMR Report
`Number 03–202 R (CMR International Surrey, UK, 2003).
`
`5.
`
`6.
`
`7.
`
`1.
`
`2.
`
`3.
`
`4.
`
`P E R S P E C T I V E S
`
`8. Van den Haak, M. A., Palachandran, J. & Benjamin, G. A.
`Performance Metrics in Global Pharmaceutical R&D,
`Company specific review 2002 for Merck & Co. (CMR
`International, Surrey, UK, 2003).
`9. Roberds, S. L. et al. BACE knockout mice are healthy
`despite lacking the primary β-secretase activity in brain:
`implications for Alzheimer’s disease therapeutics.
`Hum. Mol. Genet. 10, 1317–1324 (2001).
`10. Gowen, et al. Cathepsin K knockout mice develop
`osteopetrosis due to a deficit in matrix degradation but
`not demineralization. J. Bone Miner. Res. 14,
`1654–1663 (1999).
`11. Lazner, F., Gowen, M. & Kola, I. An animal model for
`pycnodysostosis: the role of cathepsin K in bone
`remodeling. Mol. Med. Today 5, 413–414 (1999).
`12. Ma, P. & Zemmel, R. Value of novelty? Nature Rev. Drug
`Discov. 1, 571–572 (2002).
`
`Acknowledgements
`We wish to acknowledge Datamonitor for the assembly of data
`(Pharmaceutical R&D Benchmarking Forum) used in this study.
`
`Competing interests statement
`The authors declare competing financial interests: see Web version
`for details..
`
`Online links
`
`FURTHER INFORMATION
`PhRMA: http://www.phrma.org/
`Access to this interactive links box is free online.
`
`NATURE REVIEWS | DRUG DISCOVERY
`
`VOLUME 3 | AUGUST 2004 | 7 1 5
`
`NOVARTIS EXHIBIT 2064
`Breckenridge v. Novartis, IPR 2017-01592
`Page 5 of 5
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket