throbber
V O L U M E 2 5 䡠 N U M B E R 2 䡠 J A N U A R Y 1 0 2 0 0 7
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`R E V I E W A R T I C L E
`
`From the Tufts Center for the Study of
`Drug Development, Tufts University,
`Boston, MA; Department of Econom-
`ics, Duke University, Durham, NC.
`
`Submitted September 5, 2006; accepted
`September 26, 2006.
`
`Authors’ disclosures of potential con-
`flicts of interest and author contribu-
`tions are found at the end of this
`article.
`
`Address reprint requests to Joseph A.
`DiMasi, PhD, Tufts Center for the
`Study of Drug Development, Tufts
`University, 192 South St, Suite 550,
`Boston, MA 02111; e-mail:
`joseph.dimasi@tufts.edu.
`
`© 2007 by American Society of Clinical
`Oncology
`
`0732-183X/07/2502-209/$20.00
`
`DOI: 10.1200/JCO.2006.09.0803
`
`Economics of New Oncology Drug Development
`Joseph A. DiMasi and Henry G. Grabowski
`
`A
`
`B
`
`S
`
`T
`
`R
`
`A
`
`C
`
`T
`
`Purpose
`Review existing studies and provide new results on the development, regulatory, and market
`aspects of new oncology drug development.
`Methods
`We utilized data from the US Food and Drug Administration (FDA), company surveys, and publicly
`available commercial business intelligence databases on new oncology drugs approved in the
`United States and on investigational oncology drugs to estimate average development and
`regulatory approval times, clinical approval success rates, first-in-class status, and global
`market diffusion.
`Results
`We found that approved new oncology drugs to have a disproportionately high share of FDA
`priority review ratings, of orphan drug designations at approval, and of drugs that were granted
`inclusion in at least one of the FDA’s expedited access programs. US regulatory approval times
`were shorter, on average, for oncology drugs (0.5 years), but US clinical development times were
`longer on average (1.5 years). Clinical approval success rates were similar for oncology and other
`drugs, but proportionately more of the oncology failures reached expensive late-stage clinical
`testing before being abandoned. In relation to other drugs, new oncology drug approvals were
`more often first-in-class and diffused more widely across important international markets.
`Conclusion
`The market success of oncology drugs has induced a substantial amount of investment in
`oncology drug development in the last decade or so. However, given the great need for further
`progress, the extent to which efforts to develop new oncology drugs will grow depends on future
`public-sector investment in basic research, developments in translational medicine, and regulatory
`reforms that advance drug-development science.
`
`J Clin Oncol 25:209-216. © 2007 by American Society of Clinical Oncology
`
`INTRODUCTION
`
`Although progress has been made in treating many
`forms of cancer, there remains a strong medical need
`for substantial improvement. This makes the com-
`plex economics of new oncology drug development
`an important area to research. In recent years, rising
`prices and growing expenditures on oncology
`drugs1 have caused significant concern among pay-
`ers and patients.2 At the same time, and likely due in
`part to expanded market opportunities, some data
`indicate that the development of new, often tar-
`geted, oncology therapies has recently been growing
`significantly.3,4 The extent to which markets will
`grow in the future, however, is uncertain because
`sponsors may face increasing resistance to what are
`perceived to be high and unsustainable prices, in-
`creasing competition if a substantial number of new
`therapies enter the market, and smaller market sizes
`for highly targeted therapies.4
`
`Incentives to develop new therapies also de-
`pend on the costs, risks, and length of new drug
`development. Pharmaceutical research and devel-
`opment (R&D) costs in general have been estimated
`to be high and rising substantially over time.5 Costs
`(at least clinical phase expenditures) have also been
`shown to differ by therapeutic class.6 Unfortunately,
`to date, not enough information has been available
`to reliably estimate R&D costs for oncology drugs. A
`good deal of information, however, can be gathered
`on other metrics of the drug development process
`for oncology drugs. This article will review informa-
`tion on the markets for new oncology drugs and
`present new data on the length and risks of new
`oncology drug development.
`
`METHODS
`
`To analyze various aspects of the development, regulatory,
`and market characteristics of new oncology drugs, we
`
`209
`
`Genome Ex. 1027
`Page 1 of 8
`
`

`

`DiMasi and Grabowski
`
`utilized a variety of data sources. Information on new drug US clinical devel-
`opment and approval times were obtained from public sources and company
`surveys, and were complied for a Tufts Center for the Study of Drug develop-
`ment (CSDD) database. The US clinical phase is defined here as the time from
`first filing of an investigational new drug application (IND) with the US Food
`and Drug Administration (FDA) to study a new drug in humans to first
`submission to the FDA of a new drug application (NDA) or biologic license
`application (BLA) for marketing approval of the new drug. The approval phase
`is the time from first submission of an NDA or BLA to approval of the
`application for marketing. With regard to these development and approval
`times, we focus attention on therapeutic drugs and biologics that had first
`obtained FDA approval for US marketing from 1990 through 2005. We exam-
`ined both new chemical entities and therapeutically significant new biologics.
`For the sake of brevity in expression, we refer to all of these compounds as new
`drugs. We have public NDA/BLA submission and approval dates for all of
`these new drugs, and dates of first IND filing (which have been validated with
`the FDA) for 95% of these compounds.
`We analyzed clinical approval success rates based on information ob-
`tained from a publicly available business intelligence database (IMS Health’s
`R&D Focus) for the 20 largest pharmaceutical firms in terms of pharmaceutical
`sales in 2005, with supplementary information from other commercial busi-
`ness intelligence databases. Given the lengthy development process, only com-
`pounds that had entered clinical testing through 2002 were included in the
`phase transition probability analyses. Their status was tracked through the first
`half of 2006. In addition, because a relatively large share of the compounds that
`initiated clinical testing during the latter half of the success rate analysis period
`are still active, a separate analysis for the 1998 through 2002 period would be
`questionable. Instead, to obtain a sense for the direction and extent of changes
`over time we compared results for the entire 1993 to 2002 period with results
`for the 1993 to 1997 period.
`A number of studies of drug industry success rates have used statistical
`inference techniques (mainly survival analysis) to account for the right-
`censoring of the data.5,11 However, given the relatively recent experience of the
`compounds we considered here and the length of the development process
`for many drugs, a significant number of compounds that we examined had
`not yet reached their final fate (abandonment or marketing approval),
`thereby making these statistical approaches somewhat unreliable. There-
`fore, we estimate success and phase attrition rates in a mechanistic manner.
`Specifically, we calculated phase transition probabilities by dividing the
`number of molecules that completed a given phase and entered the next
`phase by the difference between the number of molecules that entered the
`phase and those still in the phase at the time of the analysis. Such an
`approach should provide reasonable estimates of phase transition proba-
`bilities because the lengths of individual phases are short relative to total
`development times. The accuracy depends on an implicit assumption that
`those drugs that are still active at the time of analysis will proceed to later
`phases more or less in the same proportions as the estimated transition
`probabilities. The overall clinical success rate is then determined as the
`product of the phase transition probabilities. Clinical success is defined as
`US regulatory approval for marketing.
`Data on market and other characteristics of new drug launches were
`obtained from IMS Health’s New Product Focus database used for a study of
`the quality and quantity of worldwide new drug introductions.12 This
`database reports drug launches in 68 countries since 1982. The data exam-
`ined includes new biologic products, but it excludes diagnostic tests (ex-
`cept for radiopaques), radiologicals, over-the-counter drugs, combination
`vaccines, polyclonal antibodies, and biologic extracts. Launch dates were
`used to determine whether a new drug launch was for a first-in-class drug.
`Therapeutic classes for this analysis were chosen based on a unique com-
`bination of the four-digit level Anatomic Therapeutic Classification (ATC)
`and five-digit level Uniform System of Classification (USC) codes. The
`ATC and USC system are the same for many therapeutic classes, but when
`they differed, as a general principle the most disaggregate class from these
`two sources was used.
`
`RESULTS
`
`We first examined the number and regulatory characteristics of new
`oncology drug approvals in the Unites States since 1990. Table 1 lists
`the 68 new oncology drugs approved for marketing in the United
`States from 1990 to 2005, along with their NDA/BLA submission and
`approval dates. The FDA also approved 434 other new drugs (as
`defined herein) during this period. Seventy-nine percent of the ap-
`proved new oncology drugs are traditional small-molecule com-
`pounds (78% of the other new drugs approved during the study
`period are also small molecules). If we narrow the focus on large-
`molecule approvals to the most common types of approved “biotech”
`products (recombinant proteins and monoclonal antibodies [mAbs];
`excluding, for example, purified biologics), we find that 18% of the
`oncology drug approvals and 15% of the other drug approvals are
`biotech products under this definition. The biotech share of all drug
`approvals increased over time for both oncology and other drugs,
`although the rate of increase was faster for oncology drugs. The bio-
`tech shares were 8% and 9% during 1990 to 1993 for oncology and
`other drugs, respectively. However, the biotech shares rose to 29% and
`24% during 2002 to 2005 for oncology and other drugs, respectively.
`From a regulatory perspective, the oncology drugs differ mark-
`edly from other new drug approvals. As Table 2 indicates, 71% of the
`oncology drug approvals were given a priority review rating by the
`FDA, in contrast to 40% for other new drugs. Nearly half of the on-
`cology drugs were initially approved with an orphan drug indication,
`while less than one in five other drugs had orphan drug status at first
`approval. Finally, sponsors of oncology drugs were much more often
`able to take advantage of at least one of the FDA’s programs to speed
`development (subpart E, accelerated approval, fast track). Close to half
`of the approved oncology drugs had some expedited access status
`during development, as opposed to only 13% for the other new drugs
`approved during the study period.
`Oncology Drug Development Times
`As noted, oncology drugs are disproportionately given priority
`ratings by the FDA, which carries with it a performance goal for faster
`review of marketing applications. This is reflected in the approval
`phase means shown in Figure 1. The FDA reviewed oncology drugs,
`on average, 6 months faster than other drugs. We also noted that
`oncology drugs were more likely to be able to take advantage of FDA
`expedited access programs during development. However, despite
`this fact, difficulties in recruiting patients and longer times needed to
`establish efficacy (particularly if survival is an end point) for oncology
`drug clinical trials can help explain why we found US clinical develop-
`ment times to be a year and a half longer for oncology drugs. For the
`period analyzed, oncology drugs took, on average, 1 year longer to
`move from the initiation of clinical testing in the United States to US
`regulatory marketing approval. Development and approval phase
`times are lower for medians, but the comparative results are similar.
`Median approval phase times are 0.3 years shorter for oncology drugs
`(1.0 v 1.3 years), whereas median clinical phase times are 1.5 years
`longer for oncology drugs (7.8 v 6.3 years).
`Technical Success Rates for Oncology
`Drug Development
`To examine technical success rates and phase transition rates
`for investigational oncology and other drugs, we obtained data
`on the pipelines of the 20 pharmaceutical firms with the most
`
`210
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`Genome Ex. 1027
`Page 2 of 8
`
`

`

`New Oncology Drug Development
`
`Table 1. New Oncology Compounds Approved in the United States, 1990-2005
`NDA Submission
`Date
`
`Trade Name
`
`Sponsor
`
`Plenaxis
`Proleukin
`Campath
`Uroxatral
`Panretin
`Hexalen
`Ethyol
`Levulan Kerastick
`Arimidex
`Emend
`Trisenox
`Vidaza
`Pacis
`Avastin
`Targretin
`Casodex
`Velcade
`Xeloda
`Erbitux
`Leustatin
`Clolar
`Ontak
`Zinecard
`Taxotere
`Anzemet
`Avodart
`Ellence
`Tarceva
`Aromasin
`Proscar
`Fludara
`Faslodex
`Iressa
`Gemzar
`Mylotarg
`Kytril
`Zevalin
`Idamycin
`Gleevec
`Camptosar
`Revlimid
`Femara
`Ergamisol
`Actinex
`Arranon
`Nilandron
`Eloxatin
`Taxol
`Kepivance
`Aloxi
`Oncospar
`Alimta
`Nipent
`Photofrin
`Elitek
`Rituxan
`Quadramet
`Nexavar
`Temodar
`Vumon
`Hycamtin
`Fareston
`Bexxar
`Herceptin
`Trelstar Depot
`Valstar
`Navelbine
`Zometa
`
`Praecis
`Chiron
`Berlex
`Sanofi-Synthelabo
`Ligand
`U.S. Bioscience
`U.S. Bioscience
`Dusa
`Zeneca
`Merck
`Cell Therapeutics
`Pharmion
`Biochem Pharma
`Genentech
`Ligand
`Zeneca
`Millennium
`Roche
`Imclone
`Ortho
`Genzyme
`Ligand Pharmaceuticals
`Pharmacia
`Rhone-Poulenc Rorer
`Hoechst Marion Roussel
`Glaxo Wellcome
`Pharmacia & Upjohn
`Osi/Genentech
`Pharmacia & Upjohn
`Merck
`Berlex
`Astrazeneca
`Astrazeneca
`Lilly
`Wyeth-Ayerst
`Smithkline Beecham
`Idec
`Adria Labs
`Novartis
`Pharmacia & Upjohn
`Celgene
`Novartis
`Janssen
`Chemex/Reed & Carnick
`Glaxosmithkline
`Hoechst Marion Roussel
`Sanofi
`Bristol-Myers Squibb
`Amgen
`Helsinn Healthcare
`Enzon
`Eli Lilly
`Warner-Lambert
`Qlt
`Sanofi-Synthelabo
`Genentech
`Cytogen
`Bayer/Onyx
`Schering-Plough
`Bristol-Myers Squibb
`Smithkline Beecham
`Orion/Schering
`Corixa
`Genentech
`Pharmacia
`Anthra Pharmaceuticals
`Burroughs Wellcome
`Novartis
`
`12/12/2000
`12/1/1988
`12/23/1999
`12/8/2000
`5/27/1998
`12/19/1988
`9/30/1991
`7/1/1998
`3/29/1995
`9/27/2002
`3/28/2000
`12/29/2003
`4/21/1995
`9/30/2003
`6/23/1999
`9/14/1994
`1/21/2003
`10/31/1997
`8/14/2003
`12/31/1991
`3/30/2004
`12/9/1997
`2/10/1992
`7/27/1994
`9/29/1995
`12/21/2000
`12/15/1998
`7/30/2004
`12/21/1998
`4/15/1991
`11/24/1989
`3/28/2001
`8/5/2002
`2/2/1995
`10/29/1999
`4/14/1992
`11/1/2000
`8/31/1989
`2/27/2001
`12/28/1995
`4/7/2005
`7/25/1996
`11/1/1989
`4/10/1989
`4/29/2005
`3/7/1994
`6/24/2002
`7/22/1992
`6/24/2004
`9/27/2002
`1/1/1991
`9/30/2003
`2/11/1991
`4/13/1994
`12/16/1999
`2/28/1997
`6/13/1995
`7/8/2005
`8/13/1998
`9/28/1990
`12/22/1995
`1/3/1995
`9/15/2000
`5/4/1998
`6/26/1996
`12/31/1997
`8/27/1993
`12/21/1999
`
`NDA Approval
`Date
`
`11/25/2003
`5/5/1992
`5/7/2001
`6/12/2003
`2/2/1999
`12/26/1990
`12/8/1995
`12/3/1999
`12/27/1995
`3/26/2003
`9/25/2000
`5/19/2004
`3/9/2000
`2/26/2004
`12/29/1999
`10/4/1995
`5/13/2003
`4/30/1998
`2/12/2004
`2/26/1993
`12/28/2004
`2/5/1999
`5/26/1995
`5/14/1996
`9/11/1997
`11/20/2001
`9/15/1999
`11/18/2004
`10/21/1999
`6/19/1992
`4/18/1991
`4/25/2002
`5/5/2003
`5/15/1996
`5/17/2000
`12/29/1993
`2/19/2002
`9/27/1990
`5/10/2001
`6/14/1996
`12/27/2005
`7/25/1997
`6/18/1990
`9/4/1992
`10/28/2005
`9/19/1996
`8/9/2002
`12/29/1992
`12/15/2004
`7/25/2003
`2/1/1994
`2/4/2004
`10/11/1991
`12/27/1995
`7/12/2002
`11/26/1997
`3/28/1997
`12/20/2005
`8/11/1999
`7/14/1992
`5/28/1996
`5/29/1997
`6/27/2003
`9/25/1998
`6/15/2000
`9/25/1998
`12/23/1994
`8/20/2001
`
`Generic Name
`
`Abarelix
`Aldesleukin
`Alemtuzumab
`Alfuzosin
`Alitretinoin
`Altretamine
`Amifostine
`Aminolevulinic acid
`Anastrozole
`Aprepitant
`Arsenic trioxide
`Azacitidine
`Bcg, live
`Bevacizumab
`Bexarotene
`Bicalutamide
`Bortezomib
`Capecitabine
`Cetuximab
`Cladribine
`Clofarabine
`Denileukin diftotox
`Dexrazoxane
`Docetaxel
`Dolasetron mesylate
`Dutasteride
`Epirubicin
`Erlotinib
`Exemestane
`Finasteride
`Fludarabine phosphate
`Fulvestrant
`Gefitinib
`Gemcitabine hydrochloride
`Gemtuzumab ozogamicin
`Granisetron hydrochloride
`Ibritumomab tiuxetan
`Idarubicin hydrochloride
`Imatinib mesylate
`Irinotecan hydrochloride
`Lenalidomide
`Letrozole
`Levamisole hydrochloride
`Masoprocol cream, 10%
`Nelarabine
`Nilutamide
`Oxaliplatin
`Paclitaxel
`Palifermin (kgf)
`Palonosetron
`Pegaspargase
`Pemetrexed
`Pentostatin
`Porfimer
`Rasburicase
`Rituximab
`Samarium sm 153 lexidronam
`Sorafenib
`Temozolomide
`Teniposide
`Topotecan hydrochloride
`Toremifene citrate
`Tositumomab-i131
`Trastuzumab
`Triptorelin pamoate
`Valrubicin
`Vinorelbine tartrate
`Zoledronic acid
`
`Abbreviation: NDA, new drug application.
`
`www.jco.org
`
`211
`
`Genome Ex. 1027
`Page 3 of 8
`
`

`

`DiMasi and Grabowski
`
`76.8
`
`67.1
`
`57.5
`
`59.4
`
`57.1
`
`50.0
`
`1993-1997
`1993-2002
`
`26.1
`
`19.3
`
`Transition Probability (%)
`
`Table 2. Regulatory Characteristics of New Therapeutic Oncology and
`Other Drugs Approved in the United States, 1990-2005
`
`%
`
`Characteristic
`
`Oncology Drugs
`
`Other Drugs
`
`FDA priority ratingⴱ
`Orphan drug designation
`Expedited access†
`
`70.9
`48.5
`47.1
`
`40.2
`18.5
`13.4
`
`ⴱTherapeutic new molecular entities approved by FDA’s Center for Drug
`Evaluation and Research (CDER).
`†Drugs that were developed under at least one of the following three FDA
`regulatory mechanisms: subpart E, accelerated approval, fast track.
`
`pharmaceutical sales in 2005. We were able to identify 838 drugs that
`had entered the clinical testing pipeline for the first time anywhere in
`the world from 1993 to 2002. Of these drugs, 175 (21%) were investi-
`gated for oncology indications. A somewhat higher proportion of the
`investigational oncology drugs are large molecules (28%) than is the
`case for the approved drugs noted herein. The oncology drugs tended
`to be investigated for more indications than was the case for other
`investigational drugs. Whereas 46% of other investigational drugs
`were tested for more than one indication before an approval for
`marketing, 57% of the oncology drugs were investigated for mul-
`tiple indications. More notably, nearly one third of the oncology
`drugs (32%) were tested in at least four indications, whereas only
`9% of the other drugs were examined in four or more indications
`before an original approval for marketing.
`Figure 2 shows estimated clinical phase transition probabilities
`for investigational oncology drugs that first entered clinical testing
`from 1993 to 1997 and 1993 to 2002. The results indicate that one half
`of the oncology drugs that entered the expensive phase III clinical
`testing phase never make it to US regulatory approval (although, the
`approval rate is somewhat higher when the longer timeframe for drugs
`entering clinical testing is considered). The product of the phase tran-
`sition probability estimates yields an estimate of the clinical approval
`success rate for drugs entering the clinical testing pipeline. The results
`suggest that approximately one in five of the oncology drugs that
`entered the pipeline during 1993 to 1997 will eventually attain mar-
`keting approval, while the estimate improves to approximately one in
`four for the longer 1993 to 2002 period.
`
`Phase I-II
`
`Phase II-III Phase III-NDA
`Approval
`
`Phase I-NDA
`Approval
`
`Fig 2. Clinical phase transition probabilities for investigational oncology com-
`pounds for the 20 largest firms by pharmaceutical sales (2005) by period during
`which compound first entered clinical testing. NDA, new drug application.
`
`The results in Figure 2 are for all oncology drugs that were in the
`firms’ clinical testing pipelines at some point. The data include com-
`pounds that were licensed in at some point in development by one of
`the firms and a smaller proportion of drugs that these firms licensed
`out to firms outside of the group of 20. Drugs that are licensed may
`have somewhat higher success rates than those that are developed
`entirely under the auspices of a given firm (self-originated) because of
`due diligence prescreening and because they tend to be licensed after
`the drugs had progressed to later clinical phases. Figure 3 shows
`estimates of phase transition probabilities and the overall clinical
`approval success rate for self-originated oncology drugs compared
`with the results for all oncology drugs. The self-originated com-
`pounds have a slightly lower approval success rate than is the case
`for all oncology drugs.
`Finally, we examined transition probability and success rate
`results for oncology drugs compared with all other drugs. The
`results in Figure 4 cover all drugs for the entire 1993 to 2002 period.
`Oncology drugs have a higher likelihood of progressing to later
`clinical phases, but the success rate once drugs reach expensive
`phase III testing is notably lower for oncology drugs. Overall,
`though, the approval success rates for drugs entering the clinical
`testing pipeline are essentially the same.
`
`76.8
`
`70.7
`
`69.2
`
`57.1
`
`59.4
`
`50.0
`
`Self-originated
`All
`
`24.5
`
`26.1
`
`Transition Probability (%)
`
`Other drugs
`Oncology drugs
`
`7.8
`
`8.1
`
`9.1
`
`6.3
`
`1.8
`
`1.3
`
`Years
`
`Approval Phase
`
`Clinical Phase
`
`Total
`
`Phase I-II
`
`Phase II-III Phase III-NDA
`Approval
`
`Phase I-NDA
`Approval
`
`Fig 1. Mean clinical development and regulatory approval times for new
`oncology and other therapeutic molecular entities approved by the US Food and
`Drug Administration from 1990 to 2005.
`
`Fig 3. Clinical phase transition probabilities for investigational oncology com-
`pounds for the 20 largest firms by pharmaceutical sales (2005) by source of
`compound. NDA, new drug application.
`
`212
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`Genome Ex. 1027
`Page 4 of 8
`
`

`

`New Oncology Drug Development
`
`involving new and established drugs.14 Significant drug progress oc-
`curs both by introduction of novel new classes and by the evolution of
`products in these classes after the first mover is introduced.15 How-
`ever, first-in-class drug introductions represent important milestones
`in documenting the extent of drug innovation over time.
`A second major finding of the Grabowski and Wang12 analysis is
`the increasing global character of new drug introductions. Grabowski
`and Wang found that nearly half (47%) of all 1993 to 2003 new drug
`introductions were launched in a majority of the G7 countries. (The
`G7 countries were chosen as a relevant benchmark because they con-
`stitute the largest seven drug markets in terms of sales. These countries
`are the United States, Japan, the United Kingdom, Germany, France,
`Italy, and Canada.) This compares to 37% for the 1982 to 1992 period.
`Furthermore, a prior study of new drug introductions for the 1970 to
`1983 period found that only 24% of new drugs were characterized as
`global entities.16
`Grabowski and Wang also found that biotech drugs account for a
`rising portion of all new drugs over the 1982 to 2003 period. The rapid
`growth of biotech compounds is reflected in the fact that biotech drugs
`accounted for only 4% of worldwide introductions in the period 1982
`to 1992, but this increased to 16% in the 1993 to 2003 period. Further-
`more, more than half of these biotech compounds originated in US
`firms. The growth of biotech drugs is particularly significant because
`they have been a major source of both first-in-class and global drugs.
`They also have a strong presence in the oncology class.
`In this review article, we are particularly interested in how oncol-
`ogy drugs compare with other therapeutic classes with respect to these
`drug industry attributes considered in the Grabowski and Wang anal-
`ysis. In this regard, Table 3 provides a breakdown of the distribution of
`new drugs by therapeutic areas and various subcategories using
`Grabowski and Wang’s sample of 919 worldwide introductions for the
`1992 to 2003 period. All therapeutic areas with 5% or more of the total
`number of new drug introductions total are listed separately. The
`remaining areas with small numbers of introductions are combined
`into the miscellaneous category.
`
`Table 3. Therapeutic Area Distribution of New Drugs for 1982-2003
`Worldwide New Drug Introductions8
`
`Therapeutic Area
`
`Central nervous system
`Cardiovascular system
`Systemic anti-infectives
`Oncology
`Alimentary tract and metabolism
`Musculoskeletal system
`Blood and blood-forming organs
`Respiratory system
`Dermatologicals
`Miscellaneous
`Total
`
`All New
`Drugs
`
`Global New
`Drugs
`
`First-in-
`Class New
`Drugs
`
`Biotech
`New Drugs
`
`130
`128
`127
`99
`86
`70
`59
`57
`49
`118
`
`57
`45
`62
`52
`29
`28
`24
`21
`21
`49
`
`12
`7
`12
`21
`13
`5
`9
`5
`7
`24
`
`1
`5
`6
`25
`9
`7
`15
`2
`3
`18
`
`NOTE. Worldwide introductions by year are obtained from the IMS New
`Product Focus database. A global new drug is defined as a new drug
`introduced in a majority of the G7 countries. A first-in-class new drug is
`defined as the first drug introduction in a specific five-digit Uniform System of
`Classification category or a four-digit Anatomic Therapeutic Classification
`category, based on information contained in the IMS databases. Biotech drug
`classification is based on IMS designation in its New Product Focus database.
`A few drugs are classified into more than one therapeutic area so category
`totals may not equal the sum of the specific therapeutic areas.
`
`76.8
`
`68.2
`
`59.4
`
`53.3
`
`68.4
`
`57.1
`
`Other drugs
`Oncology drugs
`
`24.9
`
`26.1
`
`Transition Probability (%)
`
`Phase I-II
`
`Phase II-III Phase III-NDA
`Approval
`
`Phase I-NDA
`Approval
`
`Fig 4. Clinical phase transition probabilities for investigational oncology and
`all compounds for the 20 largest firms by pharmaceutical sales (2005) for
`compounds that first entered clinical testing during 1993 to 2002. NDA, new
`drug application.
`
`Biotech products, particularly mAbs, have become increasingly
`prevalent in oncology investigational drug pipelines. The data for the
`20 firms examined here are too limited with regard to mAbs to
`provide reliable success rate estimates. However, for a recent anal-
`ysis of biopharmaceutical R&D costs, DiMasi and Grabowski13
`examined clinical approval success rates for 522 recombinant pro-
`teins and mAbs that first entered clinical testing from 1990 to 2003
`for what is likely either the population or something close to the
`population of such products. More than half (54%) of the mAbs in
`this data set were examined for oncology indications. The clinical
`approval success rate for the biotech products in aggregate was
`30%, but only 19% for mAbs. Further analysis of that data set
`shows that the estimated success rate for the subset of oncology
`mAbs is also 19%. The data do suggest, however, an increasing
`trend in success rates for mAbs in general.
`Market Attributes and Diffusion of Oncology Drugs:
`Comparative Trends
`In a recent article, Grabowski and Wang12 examine trends in
`various attributes of worldwide new drug introductions over the pe-
`riod 1982 to 2003. In particular, they consider trends in drug “inno-
`vativeness” as indicated by the number of first-in-class introductions.
`These are essentially new drugs with a novel mechanism of action.
`Second, they consider trends in the global diffusion of worldwide new
`drug introductions. In particular, they define a new drug as global
`when it is launched in a majority of the world’s largest drug markets.
`Global diffusion is an indicator of both commercial as well as thera-
`peutic importance. They also focus on the growth in biotech products
`and orphan drug products, two groups of products with increasing
`impact on the biopharmaceutical industry over the last two decades.
`One key finding of the Grabowski and Wang12 analysis is that the
`number of first-in-class drug introductions has been increasing over
`time. This contrasts with a downward trend in overall new drug
`introductions that has been discussed by many observers.14 This latter
`trend has been cited as evidence for the declining research productivity
`of the pharmaceutical industry in recent years. However, this view
`must be qualified by the positive trend in drug innovativeness, as
`reflected by the increasing number of first-in-class products. Of
`course, therapeutic benefits are also obtained from follow-on intro-
`ductions in a new drug class as well as by combination therapies
`
`www.jco.org
`
`213
`
`Genome Ex. 1027
`Page 5 of 8
`
`

`

`DiMasi and Grabowski
`
`Table 3 indicates that oncology was the fourth largest therapeutic
`area in terms of the number of worldwide introductions (99) behind
`the CNS, cardiovascular and systemic anti-infective categories. At the
`same time, the Table shows that oncology drugs had the most first-in-
`class and biotech drugs. It also ranked third in terms of global new
`drug introductions across all the therapeutic area categories. (The
`miscellaneous category is not included in this comparison, given that
`it’s a conglomerate of many smaller drug categories.)
`It is instructive to consider the share of oncology drugs that
`embody these various attributes compared with other major drug
`classes. Consider this information for the four largest therapeutic areas
`in Table 3: the CNS, cardiovascular, anti-infective, and oncology cat-
`egories. Oncology is particularly distinguished by the large percentage
`of its new drug introductions that were first-in-class. Over the 1992 to
`2003 period, 21% of oncology introductions were first-in-class enti-
`ties, as compared with less than 10% of the introductions in the other
`three classes. This figure also shows that more than half of all oncology
`introductions were classified as global drugs as compared with 35% to
`49% of the drugs in the other three therapeutic areas.
`The results in Table 3 demonstrate that the oncology therapeutic
`area has been a focal point for the introduction of innovative first-in-
`class compounds with a high rate of global diffusion. Oncology also
`has been an increasing focus of biotech drug R&D. As can be seen from
`the data in Table 3, 25% of oncology drug approvals are based on
`biotechnology techniques, compared with 5% or less in the three other
`major classes. This is a striking difference. Biotech products are an
`important driver of strong innovative performance observed for the
`oncology class in recent years.
`
`Orphan Drug Act and Oncology Drugs
`The oncology drug class has benefited from the passage of the
`Orphan Drug Act in 1983. The Act specifically applies to illness or
`conditions with a prevalence of less than 200,000 individuals. This Act
`created a number of incentives designed to spur R&D investment for
`rare conditions and illnesses.17,18 First, the Act instructed the FDA to
`implement new protocols to facilitate orphan drug approvals, or ad-
`vanced counseling to create a more effective R&D process. Second,
`Congress created a 50% tax credit for clinical trial expenditures for
`orphan drug designations. Third, a 7-year marketing exclusivity was
`granted for FDA-designated orphan drug indications, apart from any
`patent protection that existed on these drugs.
`These provisions have been an important catalyst for the devel-
`opment of oncology drugs for rarer forms of cancer. As noted herein,
`nearly half of the oncology drugs introduced had an orphan indication
`approved at the time of initial marketing approval. Once again, this is
`a much higher percentage than what is observed for other drugs. In an
`earlier analysis17 of the first dozen years of the US Orphan Drug Act,
`the authors found that a total of 502 approved drugs and clinical drug
`candidates obtained orphan drug designation from the FDA. The
`leading indication category was cancer with 89 drug entities (17.7% of
`all drugs that received an orphan drug designation).
`Grabowski18 has analyzed the distribution of sales for 27 orphan
`drugs introduced in the 1990 to 1994 period. While there are a few
`orphan products with large annual sales, the median orphan drug in
`this sample had peak annual worldwide sales of only $29.5 million.
`The median nonorphan introduction over this same period had peak
`global sales of $236 million.6
`
`The group of 27 orphan compounds introduced from 1990 to
`1994 included six cancer treatments (the largest indication category).
`All of these six drugs also received a priority drug rating from the FDA
`as well as orphan drug designation. (The six cancer drug introductions
`receiving orphan drug approval over the 1990-1994 period were al-
`tretamine, cladribine, fludarabine phosphate, idarubicin, pentostatin,
`and teniposide.) These six orphan therapies had peak global sales that
`ranged from $2 million to $103 million. The median and mean global
`peak sales for this set of orphan cancer treatments were $12 million
`and $27 million, respectively.
`The Orphan Drug Act clearly has been an important stimulant of
`new cancer treatments for small patient populations with correspond-
`ingly modest levels of sales. Orphan drugs can realize a positive return
`on investment with smaller sales levels than can non-orphan products
`given their very different economics. First, as discussed, they have
`much smaller up-

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