throbber
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
`
`PeterEllis
`
`Pfizer Inc., Sandwich, UK
`
`Key Words
`Drug discovery = Clinical trials : Drug approval « Drug safety
`
`Abstract
`Drug developmentis an expensive, long and high-risk busi-
`ness taking 10-15 years and is associated with a high attri-
`tion rate. It is driven by medical need, disease prevalence
`and thelikelihood of success. Drug candidate selection is an
`iterative process between chemistry and biology, refining
`the molecular properties until a compound suitable for ad-
`vancing to man is found. Typically, about one in a thousand
`synthesised compoundsis ever selected for progression to
`the clinic. Prior to administration to humans, the pharmacol-
`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 |
`in healthy volunteers to assess primarily
`pharmacokinetics, safety and toleration, phase Il ina cohort
`of patients with the target disease to establish efficacy and
`dose-response relationship and large-scale phaseIll studies
`to confirm safety and efficacy. Experience tells us that ap-
`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 andits
`benefits must outweighits risks before it will be approved
`by the regulatory agencies. Strict regulatory standards gov-
`ern the conductof pre-clinical and clinicaltrials 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 5. Karger AG, Basel
`
`Introduction
`
`Getting drugs to the market is an expensive and high-
`risk business which takes on average 10-15 years to com-
`plete. The Tufts Center for the Study of Drug Develop-
`ment announced in November2001 that the average cost
`to develop a new prescription drug was USD 802 million
`[1]. Whenthe costs offailed prospective drugs are factored
`in, the actual cost for discovering, developing and launch-
`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 compoundedby the
`high attrition rate, as it is estimated that approximately
`only 1 in 10 drugsthat enter clinicaltrials will makeit to
`the market. In a recent study, it was shownthat the aver-
`age success rate for drugs to be approvedforall therapeu-
`
`
`
`KA RG E R
`Fax +4161 306 12 34
`E-Mail karger@karger.ch
`www.karger.com
`
`© 2009S, Karger AG, Basel
`1660-21 10/09/1133-0125$26,00/0
`
`Accessible onling at:
`www.karger.com/nec
`
`Pfizer Laboratories
`Ramsgate Road
`Sandwich CTI3 YN] (UR)
`Tel, +44 1304 641 627, Fax +44 1304 652 629
`E-Mail tamimis@gmail,com
`
`MPI EXHIBIT 1047 PAGE 1
`
`MPI EXHIBIT 1047 PAGE 1
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1047, p. 1 of 7
`
`

`

`~ ~ Approval
`andpost-
`approval
`
`
`
`Hypertension
`Hyperlipidaemia
`Arthritis
`
`Asthma
`Chronic kidney disease
`Obesity
`Psychotic disorders
`Malignancy
`TypeII diabetes mellitus
`Stroke
`
`Heartfailure
`Liver cirrhosis
`Chronic obstructive
`pulmonary disease
`AIDS
`
`Gastro-oesophageal reflux
`disease
`
`Epilepsy
`Type | diabetes mellitus
`
`Fig. 1. Phases of drug development.
`
`Fig. 2. Drivers for discovering new drugs
`with examples.
`
`
`
`Diseaseprevalence
`
`Medical need
`
`
`
`Genetic storage diseases
`
`Inflammatory bowel
`disease
`Irritable bowel syndrome
`
`Cystic fibrosis
`Multiple sclerosis
`Septic shock
`Transplant rejection
`
`Medium
`ed
`
`tic areas is approximately 11%. The successrate varies be-
`tween therapeutic areas ranging from 20% for cardiovas-
`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 productin clinicaltri-
`als is essential to aid in reducing the spiralling devel-
`opmentcosts. Unfortunately, increasing costs combined
`with the high attrition rate are forcing pharmaceutical
`companies to reduce investmentin research and develop-
`ment, focussing on a more limited product portfolio.
`Drug developmentis a significant challenge. Every
`product must notonly be safe andefficacious, butits ef-
`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 whatis current-
`ly the most regulated industry in the world. Once this is
`done, approved products must appeal to global markets
`across different cultures, healthcare systems anddistri-
`bution systems.
`It is interesting to note that in a recent survey, the pub-
`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 drugsare discovered by the in-
`dustry [5].
`This minireview will address the process of drug de-
`velopmentfrom discovery through the stages of develop-
`ment up to approval and marketing(fig.1).
`
`Discovery
`
`Selecting therapeutic areas or indications to invest in
`is driven by ‘medical need’ and theprevalence ofthe dis-
`ease(fig. 2). Additionalfactors also include technical fea-
`sibility, research and developmentcosts and commercial
`considerations such as competition in the market place
`and potential market share. Evenifthese criteria are met,
`there is only a limited research and development budget
`and each new project must be prioritized against the
`companyresearch and developmentportfolio, with only
`high priority projects within the budget beingselected for
`progression. For many companies,this is typically an an-
`
`c126
`
`Nephron Clin Pract 2009;113:c125-c131
`
`Tamimi/Ellis
`
`MPI EXHIBIT 1047 PAGE 2
`
`MPI EXHIBIT 1047 PAGE 2
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1047, p. 2 of 7
`
`

`

`nual review processfor productsatall stages of develop-
`ment. This maylead to stopping a programmeeven at an
`advancedstage of development.
`Early chemical starting points have been identified
`from naturally occurring substances in plants, humansor
`animals but lead compoundsare moreoften sourced from
`targeted chemical synthesis directed to bind to the known
`structures of receptors and enzymes or from random or
`receptor-targeted high-throughputscreening [6]. This has
`become more popularin the last few yearsasit is helpful
`in accelerating drug discovery. Initial problems encoun-
`tered in the last decade have eased with improving tech-
`nology. With the advent ofmodern computer technology,
`robotics and multi-well assay plates (384 growingto 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-
`other methodoflead identification is ‘virtual screening’
`(also namedin silico screening) which is defined as the
`‘selection ofcompoundsbyevaluatingtheirdesirability in
`a computational model’ [7]. Compoundstesting positive
`in screening have their potencyandselectivity confirmed
`by in vitro biochemical or cellular assays. This is typi-
`cally followed by functional biochemical and pharmaco-
`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. Onceall preclinical testing
`has satisfied the minimum selection criteria, the com-
`pound transitions from a ‘lead’ to a ‘candidate’ and is
`nominated for progression to the clinic.
`At this stage, drug productionis scaled up to meet the
`increased compound demand, work commences on de-
`veloping a suitable formulation for clinical use (often a
`tablet is the preferred dosage form) and the candidateis
`progressed through the required toxicology testing (in-
`cluding genotoxicity, safety pharmacologyin all biologi-
`cal systems, single and multiple dose toxicity and toxico-
`kinetic studies) to enable the first in human and subse-
`quent clinical studies. Reproductive toxicology in male
`and female animals (required prior to testing in women
`of child-bearing potential) and long-term carcinogenici-
`ty testing are also prerequisites forfiling a drug approval
`request [9].
`In parallel with lead development/candidate nomina-
`tion, a key decision on when to patent the compound or
`chemicalseries is taken. Early patenting mitigates against
`competitors beating a company to a claim, but delaying
`the patent application allows for introduction of addi-
`tional data to strengthen the patent and extends thepat-
`
`ent expiry date. The patentlife is typically 25 years but as
`it takes 10-15 years to develop a drug, there could only be
`10 years remainingto sell the product and recoup the
`high developmentcosts.
`
`Phasesof Clinical Drug Development
`
`Phase I. Phase I starts with thefirst administration of
`the new medicinal product to humans. Usually this phase
`involves healthy volunteers with the exception of cyto-
`toxic drugs(e.g. oncology drugs) which get tested in pa-
`tients without the requirementto test in healthy volun-
`teersfirst. The purposeofthis stageis to evaluate the safe-
`ty, tolerability, pharmacodynamic(effect of the drug on
`the body e.g. effect on heartrate, blood pressure, electro-
`cardiogram (ECG),etc.) and pharmacokinetic (effect of
`the body on the drugi.e. absorption, distribution, metab-
`olism and excretion) effects of the tested drug. Phase I
`studies are usually conducted in dedicated phase I units
`whichare research units attachedto a general or teaching
`hospital and mannedbyresearch physicians whoarefa-
`miliar with conducting such studies. Full resuscitation fa-
`cilities are available at these units. Phase I studies require
`approval from an ethics committee andtherelevant regu-
`latory agency. In the United States, an Investigational
`New Drug (IND)application, which summarises the es-
`tablished preclinical and manufacturing information
`along with investigator guidance, mustbe in place prior
`to starting clinical trials. A pre-IND consultation pro-
`grammeis offered by the US Food and Drug Administra-
`tion (FDA)to provide guidance on the data necessary for
`the IND submission. Subjects are usually compensatedfor
`participating in these studies. Developmentof the drug
`could be stoppedifit is found thatthe half-life ofthe drug
`is too short or too longorif it has poor bioavailability.
`Similarly, if the drug is not well tolerated at effective con-
`centrationsit is dropped from development. Phase I stud-
`ies usually start with single sub-pharmacological doses
`which are escalated gradually and followed by multiple
`doses. Stoppingrules to dose escalation include severe ad-
`verse events,clinically significant ECG abnormalities and
`clinically significant laboratory abnormalities.
`Otherphase 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:c125-c131
`
`c127
`
`MPI EXHIBIT 1047 PAGE 3
`
`MPI EXHIBIT 1047 PAGE 3
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1047, p. 3 of 7
`
`

`

`Role of Translational Medicine/Biomarkers
`The American Physiological Society has defined trans-
`lational research as ‘the transfer of knowledge gained
`from basic research to new and improved methodsofpre-
`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-
`fects that provide informative links between mechanisms
`of action and clinical effectiveness [11]. Effectively apply-
`ing translational research measures to a development
`programmein phase I and phaseII resultsin earlier iden-
`tification of efficacy (or just as important, lack ofeffica-
`cy) resulting in increased overall productivity and poten-
`tially a quicker route to drug approval. There are 3 fun-
`damental classifications of biomarkers: (1) markers of
`diseasee.g. proteinuria as a biomarker of chronic kidney
`disease (CKD); (2) markers ofpharmacological activity of
`a drug e.g. inhibition of angiotensin-converting enzyme
`increases plasmalevels of angiotensin-1 and decreases
`plasmalevels of angiotensin-2; (3) surrogate biomarkers
`of efficacy e.g. using a measure of penile rigidity mea-
`sured by plethysmography (Rigiscan) as a surrogate for
`sexual intercourse. An example of a biomarker with di-
`agnostic rather than efficacy potential is neutrophil gela-
`tinase-associated lipocalin or NGAL, which serves as
`biomarkerofacute renal injury as increased levels are de-
`tected in urine and blood within hoursof kidneyinjury.
`Taking the example of proteinuria in CKD,interven-
`tions that reduce proteinuria can be potentially beneficial
`in the treatment of CKD. Therefore measuring changes
`in the biomarkerin both preclinical models (e.g. sub-total
`nephrectomy model in therat) and the clinic can be in-
`dicative of activity of a potentially new drug for treating
`that indication (i.e. slowing progression of non-diabetic
`CKD). The challengeis to use or develop a biomarkerin
`which we have confidence thatit will reflect changes in
`the importantregistrable endpoints that we will assess in
`phaseIIItrials and whichareessential to gain regulatory
`approval.
`PhaseII. Once the drug’ssafety, pharmacokinetics and
`doseselection has been established in healthy volunteers,
`the nextstep is to investigate the efficacy andsafety ofthe
`drug in the target population. For example, if a drug is
`being developedfor the treatment ofhypertension, phase
`II trials will involve investigating the drug in a hyperten-
`sive patient population. Phase II is usually divided into
`phaseIIa and phaseIIb. Phase Ila is when the drug (usu-
`ally limited to a single high/maximaltolerated doselevel)
`is tested in a small cohort (12-100) of patients; this is
`
`called the ‘proof of concept’. Phase IIb follows on from
`the proof of concept in which severaldose levels are test-
`ed in the target population (dose-ranging studies) to de-
`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 IIa
`and IIb are combined in one large study. A complete
`phase II programmecould involve several hundred pa-
`tients and can cost several million dollars.
`Withever increasing developmentcosts and expiry of
`valuable patents on major products, the pharmaceutical
`industry is compelled to develop moreefficient and cost-
`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-
`stand exposure-responserelationships and optimise dose
`selection, thus facilitating regulatory review and maxi-
`mising the commercial valueofthe drug.
`However, positive phase II data is no guarantee ofpro-
`gression to phaseIII. 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-
`ing patent life of the drug, cost of goods to produce the
`drug, potential market share and pricing and reimburse-
`ment. Onceagain,the drugwill be prioritised againstall
`other candidates in the portfolio and only if the outlook
`is favourable and the priority is within the research and
`development budgetwill it go forward.
`A successful phaseII is followed by an ‘endofphase IT’
`meeting with regulatory agencies such as the FDAto dis-
`cuss the results from phaseII 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-
`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 ofadministration for approval. Be-
`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 rangeto be tested. PhaseIII trials (usually a
`minimum of 2) can involve up to several thousands of
`patients, depending on the indication, so that an ade-
`quate database (with 90% powerto detectstatistically sig-
`nificant differences) is created to assess the efficacy and
`safety profile, in addition to enabling accurate druglabel-
`
`c128
`
`Nephron Clin Pract 2009;113:c125-c131
`
`Tamimi/Ellis
`
`MPI EXHIBIT 1047 PAGE 4
`
`MPI EXHIBIT 1047 PAGE 4
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1047, p. 4 of 7
`
`

`

`Regulatory Submission/Approval
`
`if the rate is too high, additional study centres will be re-
`cruited. This ensures adequate patient numbers for ap-
`proval, butis costly in incurring delays to the programme.
`The overall success rate of phase III is around 70% and
`dependingon the size can cost up to USD 100 million. A
`successful phaseIII is usually recognised by the financial
`markets with an impact on the sponsor’s shareprice.
`
`ling. PhaseIII trials are primarily designed and powered
`to test the hypothesisofefficacy but at the sametime, ad-
`verse events are collected to assess benefit-risk potential
`of the drug. Use of novel endpointsin phaseIII is a high-
`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 authoritiespriorto the start of phaseIII.
`Clinical studies that use mortality and morbidity end-
`points are often very large and can take several years to
`complete. Oncologyis an exception, with phase III stud-
`Oncethe phaseIII studies have completed and deliv-
`ies often limited to a few hundredpatients. In diseases in
`ered a positive outcome, compilation of the data to sub-
`which there is an established ‘gold standard’ treatment,
`mit to the regulatory agenciesstarts. This usually takes
`European regulatory authorities will require phase III
`several months and can be donebyoneregion at a time,
`e.g. in the United States, or could be doneglobally, target-
`studies to include a comparator arm to demonstrate non-
`inferiority or superiority comparedto the standardther-
`ing major regions simultaneously. Classically, the major
`apy. Efficacy can be demonstrated either by demonstrat-
`markets include the United States, the European Union
`ing superiority to placebo in placebo-controlled trials or
`and Japan. However, recently moreattention is given to
`by showing superiority to an active-control treatment.
`the ‘emerging markets’ such as Latin America, India and
`Sometimesthe new drugentity is comparedto a reference
`China, amongstothers. As for the United States, a routine
`New Drug Application ‘NDA’ can take up to 15 months
`treatment without the objective of showing superiority.
`This can be either an equivalencetrial, which showsthat
`for review. However,in cases ofparticularly high medical
`the response to treatments differs by an amount whichis
`needorin areaslacking treatments(e.g. oncology and hu-
`clinically not significant (specify upper and lower equiv-
`man immunodeficiencyvirus), an expedited review can
`alence margins), or a non-inferiority trial which has the
`be granted.If the new drugis a biologic, then a biologic
`license application “BLArather than a “NDA,is submit-
`objective of showing that the new drugis notclinically
`inferior to the comparator (only lower equivalence mar-
`ted.
`gin is specified). The choice of specified margins should
`In Europe, the sponsor submits a marketing authori-
`sation application (MAA), which could be granted either
`be clinically justified.
`Depending on the nature of the study and the end-
`underthe centralised procedure(valid for the entire com-
`points used for the indication, a “Data Safety Monitoring
`munity market) or through the mutual recognition pro-
`cess.
`Board’ (DSMB)maybe required throughoutthe conduct
`During the review by the regulatory agencies, ques-
`of the trial. This is especially so in studies that incorpo-
`rate mortality and morbidity as primary or secondary
`tions are referred back to the sponsor. To facilitate the
`review process, the sponsorwill typically establish a rap-
`endpoints. DSMB membersmustincludeaclinician with
`expertise in the disease area under investigation as well
`id response team to coordinate the responsesto the au-
`thority. Drug label negotiations take place during the re-
`as a biostatistician as a minimum. Each DSMB must have
`view process. Regulatory agencies could request post-ap-
`a charter and written operating procedures detailing
`proval studies from the drug companies to address any
`members’ responsibilities and the plan of communica-
`safety concernsthat the regulatory agencies may have. At
`tion. DSMB members mustdisclose potential conflict of
`the same time, the drug companywill have presentedits
`interest to the sponsor.
`For the sponsor, phase III trials involve a large cross-
`plansto detect, assess and report adverse events.
`Pharmacovigilance is the term used in Europe de-
`functional team which involves, amongst others, clini-
`scribing the ongoing evaluation of the safety of the drug
`cians, project management, data management, drug safe-
`in the post-marketing period; it is a requirementthat all
`ty monitoring, document management, regulatory sup-
`port and clinical quality assurance. A key consideration
`pharmaceutical companies with a post marketed product
`must comply. The drug companywill also provide peri-
`for phase III is selection of study centres to ensure appro-
`odic safety update reports on the new drugafterits ap-
`priate patient recruitment and timely completion of the
`proval. Post-marketing or safety surveillance trials are
`study. Estimationsofpatient drop-outrates are made,but
`
`Drug Development
`
`Nephron Clin Pract 2009;113:c125-c131
`
`c129
`
`MPI EXHIBIT 1047 PAGE 5
`
`MPI EXHIBIT 1047 PAGE 5
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1047, p. 5 of 7
`
`

`

`sometimesreferred 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 knownas Baycolin the United States).
`Orphan Drug Status. Pharmaceutical products devel-
`oped to treat rare diseases have beenreferred to as orphan
`drugs. The FDA Orphan DrugActspecifies the require-
`ments for granting a drug orphanstatus. The disease that
`the drug is intended for should affect less than 200,000
`people in the United States. This designation grants the
`companyfast-track review process as well as market ex-
`clusivity for a period of 7 years. In addition,it will beeli-
`gible for direct guidance from the FDAforthe design of
`a clinical plan to further develop the drug. In Europe,
`some drugs used to treat tropical diseases that are pri-
`marily found in developing countries can also be desig-
`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-
`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-
`ducted according to goodclinical practice (GCP) guide-
`lines, which are internationally required quality andsafe-
`ty standards for designing, conducting and reporting
`clinical trials. GCP-compliantclinical trials are essential
`to ensure the rights and safety of clinical trial subjects.
`These standardsare subject to inspection by regulatory
`agencies at any time; regulatory agencies havethe right to
`halt ongoing clinical studies if they have concerns that
`the studies are not GCP-compliant. Finally drug manu-
`facturing is done according to good manufacturingprac-
`tice (GMP) guidelines, which dictates the standardsfor
`manufacturing and quality control of pharmaceutical
`products. This is also subject to regulatory inspection.
`
`Lifecycle Management
`
`The drug companywill plan thelifecycle of the drug
`throughout the patent life and beyondinto the future ge-
`neric marketplace. This may include different drug deliv-
`ery systems such as prolonged release formulations ver-
`
`sus immediate release, combinations with other drugs for
`improved efficacy, as well as seeking new indications.
`Oncea newindication is confirmed, the drug company
`can apply for a supplementary new drugapplication (s-
`NDA). Publication strategies are also another important
`partoflifecycle management,as additionalbenefits ofthe
`drug that cannot be addedto thelabel, such as patient-
`reported outcome measures, are published in peer-re-
`viewed journals.
`
`Interaction between Pharmaceutical Industry and
`Healthcare Professionals
`
`The Pharmaceutical Research and Manufacturers of
`America (PhRMA) represent research-based pharma-
`ceutical and biotechnology companies. PhRMAhavede-
`veloped guidelines on the basis of interactions between
`US healthcare professionals and the pharmaceutical in-
`dustry. The PhRMAcodewaslast updated in January
`2009 and regulates amongst other things: informational
`presentations by pharmaceutical company representa-
`tives and accompanying meals, prohibition on entertain-
`ment and recreation, pharmaceutical company support
`for continuing medical education, pharmaceutical com-
`pany support for third-party educationalor professional
`meetings, the employmentof healthcare professionals as
`consultants, speaker programmesand speaker training
`meetings, prohibition of non-educational and practice-
`related items as well as scholarships and educational
`funds. In the United Kingdom, the Association of the
`British Pharmaceutical Industry (ABPI) code wasestab-
`lished in 1958 and covers advertising, activities of repre-
`sentatives, supply of samples, provision of hospitality,
`promotional meetings and the sponsorship ofscientific
`and other meetings, including paymentoftravelling and
`accommodation expenses. The ABPI code doesnot apply
`to the promotion of over-the-counter medicines to the
`general public [12].
`
`Conclusion
`
`Drug developmentis a long, expensive and highly reg-
`ulated process. Therisks are high, but continued invest-
`ment in pharmaceuticals is vital if we are to enjoy the
`benefits of long-term improvements in patient health-
`care.
`
`c130
`
`Nephron Clin Pract 2009;113:c125-c131
`
`Tamimi/Ellis
`
`MPI EXHIBIT 1047 PAGE 6
`
`MPI EXHIBIT 1047 PAGE 6
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1047, p. 6 of 7
`
`

`

`5 Proteccién de la propiedad intelectual para
`productos farmacéuticos: qué es y por qué es
`esencial para la innovacién en salud? Con-
`sideraciones a la luz del DR-CAFTA.http://
`www.amchamsal.com/uploaded/content/
`category/2000065261.pdf (accessed January
`2, 2009).
`Dutta A: Discovery of new medicines; in
`Griffin JP and O’GradyJ (eds): The Textbook
`of Pharmaceutical Medicine. London, BMJ
`Books, 2002, p 25.
`International Union of Pure and Applied
`Chemistry: Glossary of terms used in com-
`binatorial chemistry, U-Z3. Research Trian-
`gle Park, International Union of Pure and
`Applied Chemistry. 1999. http://www.iupac.
`org/reports/1999/7112maclean/u-z.html
`(accessed January2, 2009).
`
`Rester U: From virtuality to reality — virtual
`screening in lead discovery and lead optimi-
`zation: a medicinal chemistry perspective.
`Curr Opin Drug Discov Devel 2008;11:559-
`568.
`
`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.
`Hall JE: The promiseof translational physi-
`ology. Am J Physiol 2002;283:1235-1236.
`Biomarkers Definitions Working Group:
`Biomarkers and surrogate endpoints: pre-
`ferred definitions and conceptual frame-
`work. Clin Pharmacol Ther 2001;69:89-95,
`What is the code of practice. http://www.
`pmcpa.org.uk/?q=whatisthecodeofpractice
`(accessed March 29, 2009).
`
`10
`
`11
`
`12
`
`References
`
`Tufts Center for the Study of Drug Develop-
`ment pegs cost of a new prescription medi-
`cine at $802 million. 2001. http://csdd.tufts.
`edu/NewsEvents/RecentNews.asp?newsid=6
`(accessed January 2, 2009).
`Drug development: the short story 7. Cost
`of drug development. Network Science Cor-
`poration. —_http://www.netsci.org/scgi-bin/
`Courseware/projector.pl?Course_num=cour-
`sel&Filename=slide07.html (accessed Janu-
`ary 2, 2009).
`Drugresearch.com: drug development costs
`hit $1.7 billion. 2003. http://www.drugre-
`searcher.com/Research-management/Drug-
`development-costs-hit-1.7-billion (accessed
`January 2, 2009).
`Kola I, Landis J: Can the pharmaceutical in-
`dustry reduceattrition rates? Nat Rev Drug
`Discov 2004;3:711-715.
`
`Editorial Comment
`
`M. El Nahas, Sheffield
`
`This minireview by TamimiandEllis, 2 senior execu-
`tives at Pfizer UK with considerable experience in drug
`development and with a genuineinterest 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 makeit to
`the bedside. Giving the cost associated with drug develop-
`mentand the currentglobalfinancial situation, this mini-
`review sheds considerable light on the direction major
`pharmaceutical companies maybe taking.First, the drug
`industry is re-evaluatingits research priorities moving to-
`wardssaferclinical areas with projected quicker financial
`return. Pfizer has moved awayfrom its prior top research
`priorities and successful drug developmentareas, namely
`atherosclerosis and heart failure research. Instead, re-
`search and developmentof drugsto tackle the growing
`market ofAlzheimer’s disease are gathering pace. Second-
`
`ly, investments in lengthyclinical trials addressing chron-
`ic diseases such as CKD mayalso fall victim to the credit
`crunch. This editor knowsof more than oneclinicaltrial
`that has been cancelled or stoppedas the sponsors’ finan-
`cial situation worsened with the global credit crunch.Fi-
`nally, drug development mayitself be shelved for a more
`cost-effective approach consisting of acquiring generic
`drug makers. Overthe last few weeks alone, Novartis pur-
`chased Ebewe Pharma an Austrian makerof generic can-
`cer drugs for USD 1.3 billion, Pfizer agreed a licencing
`deal with 2 Indian generic makers and GlaxoSmithKline
`acquired a stake in South Africa’s Aspen. Such an ap-
`proach maybe the way forward for the drug industry to
`reach a sustainable business model. It may also offer the
`industry an opportunity to unlock emerging markets that
`may accountfor 70% ofnew pharmaceuticalsales by 2020.
`Drugs development and clinical trials have not been
`spared the ravagesof the credit crunch.
`
`Drug Development
`
`Nephron Clin Pract 2009;113:c125-c131
`
`c131
`
`MPI EXHIBIT 1047 PAGE 7
`
`MPI EXHIBIT 1047 PAGE 7
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1047, p. 7 of 7
`
`

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