throbber
ne
`
`ron
`
`Clinical
`Practice
`
`Minireview
`
`Nephron Clin Pract 2009;113:c125-c131
`DOI: 10.1159/000232592
`
`Published online: August 12, 2009
`
`Drug Development: From Concept to
`Marketing!
`
`Nihad A.M. Tamimi Peter Ellis
`
`Pfizer Inc., Sandwich, UK
`
`-
`
`Key Words
`Drug discovery • Clinical trials • Drug approval • Drug safety
`
`Abstract
`Drug development is an expensive, long and high-risk busi(cid:173)
`ness taking 10- 15 years and is associated with a high attri(cid:173)
`tion rate. It is driven by medical need, disease prevalence
`and the likelihood of success. Drug candidate selection is an
`iterative process between chemistry and biology, refining
`the molecular properties until a compound suitable for ad(cid:173)
`vancing to man is found. Typically, about one in a thousand
`synthesised compounds is ever selected for progression to
`the clinic. Prior to administration to humans, the pharmacol(cid:173)
`ogy and biochemistry of the drug is established using an
`extensive range of in vitro and in vivo test procedures. It is
`also a regulatory requirement that the drug is administered
`to animals to assess its safety. Later-stage animal testing is
`also required to assess carcinogenicity and effects on the
`reproductive system. Clinical phases of drug development
`include phase I in healthy volunteers to -assess primarily
`pharmacokinetics, safety and toleration, phase II in a cohort
`of patients with the target disease to establish efficacy and
`dose-response relationship and large-scale phase Ill studies
`to confirm safety and efficacy. Experience tells us that ap(cid:173)
`proximately only 1 in 10 drugs that start the clinical phase
`will make it to the market. Each drug must demonstrate safe-
`
`ty and efficacy in the intended patient population and its
`benefits must outweigh its risks before it will be approved
`by the regul,itory agencies. Strict regulatory standards gov(cid:173)
`ern the conduct of pre-clinical and clinical trials as well as the
`manufacturing of pharmaceutical products. The assessment
`of the new medicinal product's safety continues beyond the
`initial drug approval through post-marketing monitoring of
`adverse events.
`Copyright ~ 2009 s. Karger AG, Basel
`
`Introduction
`
`Getting drugs to the market is an expensive and high(cid:173)
`risk business which takes on average 10- 15 years to com(cid:173)
`plete. The Tufts Center for the Study of Drug Develop(cid:173)
`ment announced in November 2001 that the average cost
`to develop a new prescription drug was USD 802 million
`[l]. When the costs of failed prospective drugs are factored
`in, the actual cost for discovering, developing and launch(cid:173)
`ing a single new drug would have exceeded 1.5 billion .
`This compares with USD 4 million in 1962 and USD 231
`million in 1987 [2, 3]. The problem is compounded by the
`high attrition rate, as it is estimated that approximately
`only 1 in 10 drugs that enter clinical trials will make it to
`the market. In a recent study, it was shown that the aver(cid:173)
`age success rate for drugs to be approved for all therapeu-
`
`KARGER
`l'~x +4161 106 12 ~4
`C::-Ma.il kargcr@ka1'gor.ch
`w, .. ,w.karg~r.cum
`
`I:) J0()9 S. Karger AG. Unsel
`1660-2110/09/1 133-0125S26.00/I)
`
`Acces~ible onlin4l .1.t:
`www.k.3rger.com}nt!c
`
`Pfi:r.cr Laboratories(cid:173)
`Ramsgat"e Road
`So11<lwith CTl3 9N) (UK)
`Tel. +11-1 1304 611 l 627, 1-~x. +Jl •I l J01t 652 (;29
`E-Mail lomimis@gmail.com
`
`MPI EXHIBIT 1047 PAGE 1
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1047-0001
`
`

`

`;------....---,...----..------------
`' : Discovery
`Phase Ill
`I ________ c;__.....,,.~ _ _ ...c;. _ _ _ _ ..c.. _ _ _ _ _ _ _
`
`Fig. 1. Phases of drug development.
`
`.c
`O'l
`:i:
`
`E
`:::,
`'o
`QI
`::iE
`
`;;:
`0
`....I
`
`QI
`V
`C
`QI
`io
`>
`f
`QI ..
`Q.
`QI .. 0
`.,
`
`Hypertension
`Hyperlipidaemia
`Arthritis
`
`Asthma
`Psychotic disorders
`Type II diabetes mellitus
`
`Gastro-oesophageal reflux
`disease
`
`Epilepsy
`Type I diabetes mellitus
`
`Genetic storage diseases
`
`Inflammatory bowel
`disease
`Irritable bowel syndrome
`
`Chronic kidney disease
`Obesity
`Malignancy
`Stroke
`
`Heart failure
`Liver cirrhosis
`Chronic obstructive
`pulmonary disease
`AIDS
`
`Cystic fibrosis
`Multiple sclerosis
`Septic shock
`Transplant rejection
`
`Fig. 2. Drivers for discovering new drugs
`with examples.
`
`Low
`
`Medium
`
`Medical need
`
`High
`
`tic areas is approximately 11 %. The success rate varies be(cid:173)
`tween therapeutic areas ranging from 20% for cardiovas(cid:173)
`cular drugs to only 5-8% for oncology and central nervous
`system disorder drugs [4]. Improvements in predicting
`the potential success or failure of a product in clinical tri(cid:173)
`als is essential to aid in reducing the spiralling devel(cid:173)
`opment costs. Unfortunately, increasing costs combined
`with the high attrition rate are forcing pharmaceutical
`companies to reduce investment in research and develop(cid:173)
`ment, focussing on a more limited product portfolio.
`Drug development is a significant challenge. Every
`product must not only be safe and efficacious, but its ef(cid:173)
`ficacy has also to be proven across racial and ethnic
`groups as well as across different age groups. Every drug
`has to pass a global regulatory review in what is current(cid:173)
`ly the most regulated industry in the world. Once this is
`done, approved products must appeal to global markets
`across different cultures, healthcare systems and distri(cid:173)
`bution systems.
`It is interesting to note that in a recent survey, the pub(cid:173)
`lic perception was that the pharmaceutical industry dis-
`
`covers only 27% of new drugs whilst the reality is that
`more than 90% of all new drugs are discovered by the in(cid:173)
`dustry [5].
`This minireview will address the process of drug de(cid:173)
`velopment from discovery through the stages of develop(cid:173)
`ment up to approval and marketing (fig. 1).
`
`Discovery
`
`Selecting therapeutic areas or indications to invest in
`is driven by 'medical need' and the prevalence of the dis(cid:173)
`ease (fig. 2). Additional factors also include technical fea(cid:173)
`sibility, research and development costs and commercial
`considerations such as competition in the market place
`and potential market share. Even if these criteria are met,
`there is only a limited research and development budget
`and each new project must be prioritized against the
`company research and development portfolio, with only
`high priority projects within the budget being selected for
`progression. For many companies, this is typically an an-
`
`c126
`
`Nephron Clin Pract 2009;113:cl25-cl3l
`
`Tamimi/Ellis
`
`MPI EXHIBIT 1047 PAGE 2
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1047-0002
`
`

`

`nual review process for products at all stages of develop(cid:173)
`ment. This may lead to stopping a programme even at an
`advanced stage of development.
`Early chemical starting points have been identified
`from naturally occurring substances in plants, humans or
`animals but lead compounds are more often sourced from
`targeted chemical synthesis directed to bind to the known
`structures of receptors and enzymes or from random or
`receptor-targeted high-throughput screening [6]. This has
`become more popular in the last few years as it is helpful
`in accelerating drug discovery. Initial problems encoun(cid:173)
`tered in the last decade have eased with improving tech(cid:173)
`nology. With the advent of modern computer technology,
`robotics and multi-well assay plates (384 growing to 1,536
`wells per plate), high-throughput screening can test vast
`'libraries' of chemical compounds in multiple screens
`(which can deliver up to 120,000 assays every 24 h). An(cid:173)
`other method of lead identification is 'virtual screening'
`(also named in silico screening) which is defined as the
`'selection of compounds by evaluating their desirability in
`a computational model' [7]. Compounds testing positive
`in screening have their potency and selectivity confirmed
`by in vitro biochemical or cellular assays. This is typi(cid:173)
`cally followed by functional biochemical and pharmaco(cid:173)
`logical testing in vitro, followed by pharmacodynamic
`and pharmacokinetic testing in vitro and in vivo [8]. The
`next step is to complete pilot toxicology testing to inform
`us of the likely safety profile. Once all preclinical testing
`has satisfied the minimum selection criteria, the com(cid:173)
`pound transitions from a 'lead' to a 'candidate' and is
`nominated for progression to the clinic.
`At this stage, drug production is scaled up to meet the
`increased compound demand, work commences on de(cid:173)
`veloping a suitable formulation for clinical use (often a
`tablet is the preferred dosage form) and the candidate is
`progressed through the required toxicology testing (in(cid:173)
`cluding genotoxicity, safety pharmacology in all biologi(cid:173)
`cal systems, single and multiple dose toxicity and toxico(cid:173)
`kinetic studies) to enable the first in human and subse(cid:173)
`quent clinical studies. Reproductive toxicology in male
`and female animals (required prior to testing in women
`of child-bearing potential) and long-term carcinogenici(cid:173)
`ty testing are also prerequisites for filing a drug approval
`request [9].
`In parallel with lead development/candidate nomina(cid:173)
`tion, a key decision on when to patent the compound or
`chemical series is taken. Early patenting mitigates against
`competitors beating a company to a claim, but delaying
`the patent application allows for introduction of addi(cid:173)
`tional data to strengthen the patent and extends the pat-
`
`ent expiry date. The patent life is typically 25 years but as
`it takes 10-15 years to develop a drug, there could only be
`10 years remaining to sell the product and recoup the
`high development costs.
`
`Phases of Clinical Drug Development
`
`Phase I. Phase I starts with the first administration of
`the new medicinal product to humans. Usually this phase
`involves healthy volunteers with the exception of cyto(cid:173)
`toxic drugs (e.g. oncology drugs) which get tested in pa(cid:173)
`tients without the requirement to test in healthy volun(cid:173)
`teers first. The purpose of this stage is to evaluate the safe(cid:173)
`ty, tolerability, pharmacodynamic (effect of the drug on
`the body e.g. effect on heart rate, blood pressure, electro(cid:173)
`cardiogram (ECG), etc.) and pharmacokinetic (effect of
`the body on the drug i.e. absorption, distribution, metab(cid:173)
`olism and excretion) effects of the tested drug. Phase I
`studies are usually conducted in dedicated phase I units
`which are research units attached to a general or teaching
`hospital and manned by research physicians who are fa(cid:173)
`miliar with conducting such studies. Full resuscitation fa(cid:173)
`cilities are available at these units. Phase I studies require
`approval from an ethics committee and the relevant regu(cid:173)
`latory agency. In the United States, an Investigational
`New Drug (IND) application, which summarises the es(cid:173)
`tablished preclinical and manufacturing information
`along with investigator guidance, must be in place prior
`to starting clinical trials. A pre-IND consultation pro(cid:173)
`gramme is offered by the US Food and Drug Administra(cid:173)
`tion (FDA) to provide guidance on the data necessary for
`the IND submission. Subjects are usually compensated for
`participating in these studies. Development of the drug
`could be stopped if it is found that the half-life of the drug
`is too short or too long or if it has poor bioavailability.
`Similarly, if the drug is not well tolerated at effective con(cid:173)
`centrations it is dropped from development. Phase I stud(cid:173)
`ies usually start with single sub-pharmacological doses
`which are escalated gradually and followed by multiple
`doses. Stopping rules to dose escalation include severe ad(cid:173)
`verse events, clinically significant ECG abnormalities and
`clinically significant laboratory abnormalities.
`Other phase I studies to support drug development are
`conducted throughout phase II and II of development.
`These include drug-drug interaction studies, effect of
`food on absorption, age and genetic influences. A typical
`phase I study can cost up to USD 500,000, with speciality
`studies (such as detailed QTc ECG assessments) costing
`up to USD 1.5 million.
`
`Drug Development
`
`Nephron Clin Pract 2009;113:cl25-cl31
`
`c127
`
`-
`
`MPI EXHIBIT 1047 PAGE 3
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1047-0003
`
`

`

`Role of Translational Medicine/Bio markers
`The American Physiological Society has defined trans(cid:173)
`lational research as 'the transfer of knowledge gained
`from basic research to new and improved methods of pre(cid:173)
`venting, diagnosing, or treating disease, as well as the
`transfer of clinical insights into hypotheses that can be
`tested and validated in the basic research laboratory' [10].
`Biomarkers are quantitative measures of biological ef(cid:173)
`fects that provide informative links between mechanisms
`of action and clinical effectiveness [11]. Effectively apply(cid:173)
`ing translational research measures to a development
`programme in phase I and phase II results in earlier iden(cid:173)
`tification of efficacy (or just as important, lack of effica(cid:173)
`cy) resulting in increased overall productivity and poten(cid:173)
`tially a quicker route to drug approval. There are 3 fun(cid:173)
`damental classifications of biomarkers: (1) markers of
`disease e.g. proteinuria as a biomarker of chronic kidney
`disease (CKD); (2) markers of pharmacological activity of
`a drug e.g. inhibition of angiotensin-converting enzyme
`increases plasma levels of angiotensin-1 and decreases
`plasma levels of angiotensin-2; (3) surrogate biomarkers
`of efficacy e.g. using a measure of penile rigidity mea(cid:173)
`sured by plethysmography (Rigiscan) as a surrogate for
`sexual intercourse. An example of a biomarker with di(cid:173)
`agnostic rather than efficacy potential is neutrophil gela(cid:173)
`tinase-associated lipocalin or NGAL, which serves as
`biomarker of acute renal injury as increased levels are de(cid:173)
`tected in urine and blood within hours of kidney injury.
`Taking the example of proteinuria in CKD, interven(cid:173)
`tions that reduce proteinuria can be potentially beneficial
`in the treatment of CKD. Therefore measuring changes
`in the biomarker in both preclinical models (e.g. sub-total
`nephrectomy model in the rat) and the clinic can be in(cid:173)
`dicative of activity of a potentially new drug for treating
`that indication (i.e. slowing progression of non-diabetic
`CKD). The challenge is to use or develop a biomarker in
`which we have confidence that it will reflect changes in
`the important registrable endpoints that we will assess in
`phase III trials and which are essential to gain regulatory
`approval.
`Phase II. Once the drug's safety, pharmacokinetics and
`dose selection has been established in healthy volunteers,
`the next step is to investigate the efficacy and safety of the
`drug in the target population. For example, if a drug is
`being developed for the treatment of hypertension, phase
`II trials will involve investigating the drug in a hyperten(cid:173)
`sive patient population. Phase II is usually divided into
`phase Ila and phase Ilb. Phase Ila is when the drug (usu(cid:173)
`ally limited to a single high/maximal tolerated dose level)
`is tested in a small cohort (12-100) of patients; this is
`
`called the 'proof of concept'. Phase Ilb follows on from
`the proof of concept in which several dose levels are test(cid:173)
`ed in the target population (dose-ranging studies) to de(cid:173)
`fine the minimally effective or non-effective dose and to
`decide the optimal dose, based on clinical efficacy and
`safety, to take to the next stage. Occasionally phases Ila
`and Ilb are combined in one large study. A complete
`phase II programme could involve several hundred pa(cid:173)
`tients and can cost several million dollars.
`With ever increasing development costs and expiry of
`valuable patents on major products, the pharmaceutical
`industry is compelled to develop more efficient and cost(cid:173)
`effective ways of doing drug development. These include
`the use of biomarkers, as discussed, but also application
`of enhanced quantitative drug design 'EQDD' to under(cid:173)
`stand exposure-response relationships and optimise dose
`selection, thus facilitating regulatory review and maxi(cid:173)
`mising the commercial value of the drug.
`However, positive phase II data is no guarantee of pro(cid:173)
`gression to phase III. At this key stage of development,
`costs will increase significantly and detailed analyses of
`the drug candidate and the market (patient, payer and
`physician perspectives) are conducted. This will include
`drug efficacy relative to the competitors, safety profile,
`probability of technical and regulatory success, remain(cid:173)
`ing patent life of the drug, cost of goods to produce the
`drug, potential market share and pricing and reimburse(cid:173)
`ment. Once again, the drug will be prioritised against all
`other candidates in the portfolio and only if the outlook
`is favourable and the priority is within the research and
`development budget will it go forward.
`A successful phase II is followed by an 'end of phase II'
`meeting with regulatory agencies such as the FDA to dis(cid:173)
`cuss the results from phase II and discuss and agree the
`clinical and statistical analysis plans for phase III. This
`negotiation, which also includes the target labelling, is
`critical to ensure alignment between the regulatory agen(cid:173)
`cy and sponsor.
`Phase III. This is the final stage of drug development
`prior to registration and will confirm the clinical doses,
`frequency and timing of administration for approval. Be(cid:173)
`fore embarking on a costly phase III programme, the
`sponsor should have a high level of confidence in the
`drug's safety and efficacy in the target patient population
`and the dose range to be tested. Phase III trials (usually a
`minimum of 2) can involve up to several thousands of
`patients, depending on the indication, so that an ade(cid:173)
`quate database (with 90% power to detect statistically sig(cid:173)
`nificant differences) is created to assess the efficacy and
`safety profile, in addition to enabling accurate drug label-
`
`c128
`
`Nephron Clin Pract 2009;113:cl25-cl3l
`
`Tamimi/Ellis
`
`MPI EXHIBIT 1047 PAGE 4
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1047-0004
`
`

`

`ling. Phase III trials are primarily designed and powered
`to test the hypothesis of efficacy but at the same time, ad(cid:173)
`verse events are collected to assess benefit-risk potential
`of the drug. Use of novel endpoints in phase III is a high(cid:173)
`risk strategy, but can prove valuable in demonstrating
`benefits relative to competitors or established therapies;
`however, these endpoints do require validation and
`should be included in phase II and discussed with the
`regulatory authorities prior to the start of phase III.
`Clinical studies that use mortality and morbidity end(cid:173)
`points are often very large and can take several years to
`complete. Oncology is an exception, with phase III stud(cid:173)
`ies often limited to a few hundred patients. In diseases in
`which there is an established 'gold standard' treatment,
`European regulatory authorities will require phase III
`studies to include a comparator arm to demonstrate non(cid:173)
`inferiority or superiority compared to the standard ther(cid:173)
`apy. Efficacy can be demonstrated either by demonstrat(cid:173)
`ing superiority to placebo in placebo-controlled trials or
`by showing superiority to an active-control treatment.
`Sometimes the new drug entity is compared to a reference
`treatment without the objective of showing superiority.
`This can be either an equivalence trial, which shows that
`the response to treatments differs by an amount which is
`clinically not significant (specify upper and lower equiv(cid:173)
`alence margins), or a non-inferiority trial which has the
`objective of showing that the new drug is not clinically
`inferior to the comparator (only lower equivalence mar(cid:173)
`gin is specified). The choice of specified margins should
`be clinically justified.
`Depending on the nature of the study and the end(cid:173)
`points used for the indication, a 'Data Safety Monitoring
`Board' (DSMB) may be required throughout the conduct
`of the trial. This is especially so in studies that incorpo(cid:173)
`rate mortality and morbidity as primary or secondary
`endpoints. DSMB members must include a clinician with
`expertise in the disease area under investigation as well
`as a biostatistician as a minimum. Each DSMB must have
`a charter and written operating procedures detailing
`members' responsibilities and the plan of communica(cid:173)
`tion. DSMB members must disclose potential conflict of
`interest to the sponsor.
`For the sponsor, phase III trials involve a large cross(cid:173)
`functional team which involves, amongst others, clini(cid:173)
`cians, project management, data management, drug safe(cid:173)
`ty monitoring, document management, regulatory sup(cid:173)
`port and clinical quality assurance. A key consideration
`for phase III is selection of study centres to ensure appro(cid:173)
`priate patient recruitment and timely completion of the
`study. Estimations of patient drop-out rates are made, but
`
`if the rate is too high, additional study centres will be re(cid:173)
`cruited. This ensures adequate patient numbers for ap(cid:173)
`proval, but is costly in incurring delays to the programme.
`The overall success rate of phase III is around 70% and
`depending on the size can cost up to USD 100 million. A
`successful phase III is usually recognised by the financial
`markets with an impact on the sponsor's share price.
`
`Regulatory Submission/Approval
`
`Once the phase III studies have completed and deliv(cid:173)
`ered a positive outcome, compilation of the data to sub(cid:173)
`mit to the regulatory agencies starts. This usually takes
`several months and can be done by one region at a time,
`e.g. in the United States, or could be done globally, target(cid:173)
`ing major regions simultaneously. Classically, the major
`markets include the United States, the European Union
`and Japan. However, recently more attention is given to
`the 'emerging markets' such as Latin America, India and
`China, amongst others. As for the United States, a routine
`New Drug Application 'NDA' can take up to 15 months
`for review. However, in cases of particularly high medical
`need or in areas lacking treatments (e.g. oncology and hu(cid:173)
`man immunodeficiency virus), an expedited review can
`be granted. If the new drug is a biologic, then a biologic
`license application 'BLA' rather than a 'NDA', is submit(cid:173)
`ted.
`In Europe, the sponsor submits a marketing authori(cid:173)
`sation application (MAA), which could be granted either
`under the centralised procedure (valid for the entire com(cid:173)
`munity market) or through the mutual recognition pro(cid:173)
`cess.
`During the review by the regulatory agencies, ques(cid:173)
`tions are referred back to the sponsor. To facilitate the
`review process, the sponsor will typically establish a rap(cid:173)
`id response team to coordinate the responses to the au(cid:173)
`thority. Drug label negotiations take place during the re(cid:173)
`view process. Regulatory agencies could request post-ap(cid:173)
`proval studies from the drug companies to address any
`safety concerns that the regulatory agencies may have. At
`the same time, the drug company will have presented its
`plans to detect, assess and report adverse events.
`Pharmacovigilance is the term used in Europe de(cid:173)
`scribing the ongoing evaluation of the safety of the drug
`in the post-marketing period; it is a requirement that all
`pharmaceutical companies with a post marketed product
`must comply. The drug company will also provide peri(cid:173)
`odic safety update reports on the new drug after its ap(cid:173)
`proval. Post-marketing or safety surveillance trials are
`
`Drug Development
`
`Nephron Clin Pract 2009;113:cl25-cl31
`
`c129
`
`-
`
`MPI EXHIBIT 1047 PAGE 5
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1047-0005
`
`

`

`sometimes referred to as phase IV clinical trials. Harmful
`effects discovered during phase IV trials can lead to the
`withdrawal of the drug from the market as seen in the
`example of rofecoxib (Vioxx) and cerivastatin (Lipobay,
`also known as Baycol in the United States).
`Orphan Drug Status. Pharmaceutical products devel(cid:173)
`oped to treat rare diseases have been referred to as orphan
`drugs. The FDA Orphan Drug Act specifies the require(cid:173)
`ments for granting a drug orphan status. The disease that
`the drug is intended for should affect less than 200,000
`people in the United States. This designation grants the
`company fast-track review process as well as market ex(cid:173)
`clusivity for a period of 7 years. In addition, it will be eli(cid:173)
`gible for direct guidance from the FDA for the design of
`a clinical plan to further develop the drug. In Europe,
`some drugs used to treat tropical diseases that are pri(cid:173)
`marily found in developing countries can also be desig(cid:173)
`nated as orphan drugs. For the drug companies, the cost
`of developing such drugs and marketing them will not be
`covered by the expected sales. Hence, the economic and
`regulatory incentives to encourage pharmaceutical com(cid:173)
`panies to develop such drugs are needed.
`
`Regulatory Standards
`
`Preclinical studies are conducted according to good
`laboratory practice ( GLP) guidelines, which regulate how
`laboratory studies are performed. Clinical trials are con(cid:173)
`ducted according to good clinical practice (GCP) guide(cid:173)
`lines, which are internationally required quality and safe(cid:173)
`ty standards for designing, conducting and reporting
`clinical trials. GCP-compliant clinical trials are essential
`to ensure the rights and safety of clinical trial subjects.
`These standards are subject to inspection by regulatory
`agencies at any time; regulatory agencies have the right to
`halt ongoing clinical studies if they have concerns that
`the studies are not GCP-compliant. Finally drug manu(cid:173)
`facturing is done according to good manufacturing prac(cid:173)
`tice (GMP) guidelines, which dictates the standards for
`manufacturing and quality control of pharmaceutical
`products. This is also subject to regulatory inspection.
`
`Lifecycle Management
`
`The drug company will plan the lifecycle of the drug
`throughout the patent life and beyond into the future ge(cid:173)
`neric marketplace. This may include different drug deliv(cid:173)
`ery systems such as prolonged release formulations ver-
`
`sus immediate release, combinations with other drugs for
`improved efficacy, as well as seeking new indications.
`Once a new indication is confirmed, the drug company
`can apply for a supplementary new drug application (s(cid:173)
`NDA). Publication strategies are also another important
`part oflifecycle management, as additional benefits of the
`drug that cannot be added to the label, such as patient(cid:173)
`reported outcome measures, are published in peer-re(cid:173)
`viewed journals.
`
`Interaction between Pharmaceutical Industry and
`Healthcare Professionals
`
`The Pharmaceutical Research and Manufacturers of
`America (PhRMA) represent research-based pharma(cid:173)
`ceutical and biotechnology companies. PhRMA have de(cid:173)
`veloped guidelines on the basis of interactions between
`US healthcare professionals and the pharmaceutical in(cid:173)
`dustry. The PhRMA code was last updated in January
`2009 and regulates amongst other things: informational
`presentations by pharmaceutical company representa(cid:173)
`tives and accompanying meals, prohibition on entertain(cid:173)
`ment and recreation, pharmaceutical company support
`for continuing medical education, pharmaceutical com(cid:173)
`pany support for third-party educational or professional
`meetings, the employment of healthcare professionals as
`consultants, speaker programmes and speaker training
`meetings, prohibition of non-educational and practice(cid:173)
`related items as well as scholarships and educational
`funds. In the United Kingdom, the Association of the
`British Pharmaceutical Industry (ABPI) code was estab(cid:173)
`lished in 1958 and covers advertising, activities of repre(cid:173)
`sentatives, supply of samples, provision of hospitality,
`promotional meetings and the sponsorship of scientific
`and other meetings, including payment of travelling and
`accommodation expenses. The ABPI code does not apply
`to the promotion of over-the-counter medicines to the
`general public [12].
`
`Conclusion
`
`Drug development is a long, expensive and highly reg(cid:173)
`ulated process. The risks are high, but continued invest(cid:173)
`ment in pharmaceuticals is vital if we are to enjoy the
`benefits of long-term improvements in patient health(cid:173)
`care.
`
`c130
`
`Nephron Clin Pract 2009;113:cl25-cl31
`
`Tamimi/Ellis
`
`MPI EXHIBIT 1047 PAGE 6
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1047-0006
`
`

`

`References
`
`Tufts Center for the Study of Drug Develop(cid:173)
`ment pegs cost of a new prescription medi(cid:173)
`cine at $802 million. 2001. http://csdd.tufts.
`edu/NewsEvents/RecentNews.asp?newsid=6
`(accessed January 2, 2009).
`2 Drug development: the short story 7. Cost
`of drug development. Network Science Cor(cid:173)
`poration.
`http://www.netsci.org/scgi-bin/
`Courseware/projector.pl?Course_num=cour(cid:173)
`sel&Filename=slide07.html (accessed Janu(cid:173)
`ary 2, 2009).
`3 Drugresearch.com: drug development costs
`hit $1.7 billion. 2003. http://www.drugre(cid:173)
`searcher.com/Research-management/Drug(cid:173)
`development-costs-hit-1.7-billion (accessed
`January 2, 2009).
`4 Kola I, Landis J: Can the pharmaceutical in(cid:173)
`dustry reduce attrition rates? Nat Rev Drug
`Discov 2004;3:711-715.
`
`Editorial Comment
`M. El Nahas, Sheffield
`
`5 Protecci6n de la propiedad intelectual para
`productos farmaceuticos: que es y por que es
`esencial para la innovaci6n en salud? Con(cid:173)
`sideraciones a la luz del DR-CAFTA. http://
`www.amchamsal.com/uploaded/content/
`category/2000065261.pdf (accessed January
`2, 2009).
`6 Dutta A: Discovery of new medicines; in
`Griffin JP and O'Grady J (eds): The Textbook
`of Pharmaceutical Medicine. London, BMJ
`Books, 2002, p 25.
`7 International Union of Pure and Applied
`Chemistry: Glossary of terms used in com(cid:173)
`binatorial chemistry, U-Z3. Research Trian(cid:173)
`gle Park, International Union of Pure and
`Applied Chemistry. 1999. http://www.iupac.
`org/reports/ l 999/7 l l 2maclean/u-z.html
`(accessed January 2, 2009).
`
`8 Rester U: From virtuality to reality - virtual
`screening in lead discovery and lead optimi(cid:173)
`zation: a medicinal chemistry perspective.
`Curr Opin Drug Discov Devel 2008;11:559-
`568.
`9 Tweats DJ, Scales MDC: Toxicity testing; in
`Griffin JP and O'Grady J (eds): The Textbook
`of Pharmaceutical Medicine. London, BMJ
`Books, 2002, p 134.
`10 Hall JE: The promise of translational physi(cid:173)
`ology. Am J Physiol 2002;283:1235-1236.
`11 Biomarkers Definitions Working Group:
`Biomarkers and surrogate endpoints: pre(cid:173)
`ferred definitions and conceptual frame(cid:173)
`work. Clin Pharmacol Ther 2001;69:89-95.
`12 What is the code of practice. http://www.
`pmcpa.org.uk/?q=whatisthecodeofpractice
`(accessed March 29, 2009).
`
`-
`
`This minireview by Tamimi and Ellis, 2 senior execu(cid:173)
`tives at Pfizer UK with considerable experience in drug
`development and with a genuine interest in nephrology
`and chronic kidney disease (CKD), is timely. It reminds
`the reader of the huge and often prohibitive cost of new
`drug developments. It highlights the fact that thousands
`of new potentially promising products never make it to
`the bedside. Giving the cost associated with drug develop(cid:173)
`ment and the current global financial situation, this mini(cid:173)
`review sheds considerable light on the direction major
`pharmaceutical companies may be taking. First, the drug
`industry is re-evaluating its research priorities moving to(cid:173)
`wards safer clinical areas with projected quicker financial
`return. Pfizer has moved away from its prior top research
`priorities and successful drug development areas, namely
`atherosclerosis and heart failure research. Instead, re(cid:173)
`search and development of drugs to tackle the growing
`market of Alzheimer's disease are gathering pace.

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