throbber
“Basic research is like shooting
`an arrow in the air and. where it lands.
`
`painting a target."
`
`Homer Adkins. I984
`Nature 312, 212.
`
`Food for Thought
`Look Back in Anger - What Clinical Studies
`Tell Us About Preclinical Work
`
`Thomas Hartung
`Johns Hopkins University, Bloomberg School of Public Health. CAAT, Baltimore, USA and University of Konstanz,
`CAAT-Europe, Gennany
`
`Summary
`Misled by animal studies and basic research? Whenever we take a closer look at the outcome ofclinical
`trials in afield such as, most recently, stroke or septic shock, we see how limited the value of our preclinical
`models was. For all indications, 95% of drugs that enter clinical trials do not make it to the market. despite
`all promise of the (animal) models used to develop them. Drug development has started already to decrease
`its reliance on animal models: In Europe, for example, despite intreasing R&D expenditure, animal use
`by pharmaceutical companies dropped by more than 25 % from 2005 to 2008. In vitro studies are likewise
`limited: questionable cell authenticity, over-passoging, mycoplasma infections, an.d lack ofdi}f)"erentiation
`as well as tron-homeostatic and flan-plt)’.'iir)l()giC culture conditions endanger the relevance of these models.
`The standards of statistics and reporting often are poor, further impairing reliabilit_v. Alarming studies from
`industry show miserable reproducibility of landmark studies. This paper discussesfactors contributing to
`the lack ofrepmducibility and relevanc'e ofpre-clinical research. The conclusion: Publish less but of better
`quality and do not rely on theface value of animal studies.
`
`Keywords: preclinical studies, animal studies, in vitro studies, toxicology, safety pharmacology
`
`Introduction
`
`Toxicology (Fig.1).
`
`The prime goal of biornedicine is to understand, treat, and pre-
`vent diseases. Drug development represents a key goal of re—
`Search and the pharmaceutical industry. A devastating attrition
`rate of more than 90% for substances entering clinical trials
`has received increasing attention. Obviously, we often are not
`putting our money on the right horses... Side effects not pre-
`dicted in time from toxicology and safety pharmacology con-
`tribute 20-40% to these failures, indicating limitations of the
`toolbox, which is considerably larger than what is applied to en-
`vironmental chemicals. with the exception of pesticides. Here,
`the question is raised whether quality problems of the disease
`models and basic (especially academic) research also contribute
`to this. In a simplistic view. clinical trials are based on the pillars
`of basic research/pre-clinical drug development, and toxicology
`
`What does this tell Us for areas where we have few or no clini-
`
`The temple of
`biomedica| science
`
`'
`
`,
`
`Clinical
`Studies
`
`'
`
`__ "
`I
`
`.
`
`Basic research
`
`cal trials to correct false conclusions? Toxicology is a prime ex-
`ample, where regulatory decisions for products traded at $ 10
`
`Fi9- 1: Clinical trials are based On the Piiiafs Of basic
`|'9593|‘¢h/ P|'e'Cii"iCfli Ci"-'9 d3V9i°Pm9"is and l°Xi¢°l°9V
`
`ALTEX 30, 3113
`
`Z75
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2056 - 0001
`
`

`
`HARTUNG
`
`Jgj
`
`trillion per year are taken only on the basis of such testing (Bot-
`tini and I-lartung, 2009. 2010). Are we sorting out the wrong
`candidate substances? Aspirin likely would fail the preclinical
`stage today (Hartung, 2009c). Rats and mice predict each other
`for complex endpoints with only 60% accuracy and. predicted
`together, only 43% of clinical toxicities of candidate drugs ob-
`served later (Olson et al., 2000). New approaches that rely on
`molecular pathways of human toxicity currently are emerging
`under the name “Toxicology for the 2 I 5‘ Century".
`Doubt as to animal models also is increasing: A number of in-
`creasingly systematic reviews summarized here more and more
`show the limitations. A National Academy of Sciences panel
`recently analyzed the suitability of animal models to assess the
`human efficacy of countermeasures to bioterrorism:
`It could
`neither identify suitable models nor did it recommend their de-
`velopment; it did, however, call for the establishment of other
`human-relevant tools. In line with this, about $ 200 million have
`been made available by NIH, FDA, and DoD agencies over the
`last year to start developing a human-on-a-chip approach (Har-
`tung and Zurlo, 2012).
`Academic research represents a major stimulus for drug de-
`velopment. Obviously, basic research also is carried out in phar-
`maceutical industry, but quality standards are different and the
`lesser degree of publication makes them less accessible for anal-
`ysis. Obviously, academic research comes in many flavors, and
`when pinpointing some critical notions here,each and every one
`might be unfair and not hold for a given laboratory. Similarly.
`the author and his generations of students are not free from the
`alleged (mis)behaviors. It is the far too frequent, retrospective
`view, imprinted from experiences from quality assurance and
`validation that will be shared here.
`
`Consideration 1:
`
`The crisis of drug development
`
`The situation is clear: Companies spend more and more money
`on drug development, with an average of $ 4 and up to $ 11 bil-
`lion quoted by Forbes for a successful launch to the market‘.
`The number of substances making it to market launch is drop-
`ping, and their success does not necessarily compensate for the
`increased investment. The blockbuster model of drug industry
`seems largely busted.
`The situation was characterized earlier (Hartung and Zurlo,
`2012), and more recent figures do not suggest any turn for the
`better: Failure rates in the clinical phase of development now
`reach 95% (Arrowsmjth. 2012). Analysis by the Centre for
`Medicines Research (CMR) of projects from a group of 16 com-
`panies (representing approximately 60% of global R&D spend-
`ing) in the CMR lntemational Global R&D database reveals that
`the Phase ll success rates for new development projects have
`fallen from 28% (2006-2007) to |8% (2008-2009) (Arrowsmith.
`
`2011a). 51% were due to insufficient efficacy. 29% were due
`to strategic reasons. and 19% were due to clinical or preclini-
`cal safety reasons. The average for the combined success rate
`at Phase III and submission has fallen to --50% in recent years
`(Arrowsmith. 20l lb). Taken together. clinical phases II & Ill
`now eliminate 95% of drug candidates.
`This appeared to correspond to dropping numbers of new
`drugs, as observed between I997 and 2006, as we have occa-
`sionally referenced (Bottini and Hartung. 2009. 20l0), though
`this has been shown to be possibly largely an artifact (Ward et
`al., 2013). We also have to consider that attrition does not end
`with the market launch of drugs: Unexpected side effects lead to
`withdrawals — Wikipedia, who knows it all, lists 47 drugs with-
`drawn from the market since 19903, which represents roughly
`the number of new drug entities entering the market in two years.
`This does not even include the drugs for which indications had
`to be limited because of problems. There also are examples of
`drugs that made it through the trials to the market but, in ret-
`rospect, did not work (see for examples the AP press coverage
`in October 2009 following the US Government Accountability
`Office report analyzing I44 studies, and showing that the FDA
`has never pulled a drug off the market clue to a lack of required
`follow-up about its actual benefits3).
`At the same time, combining the results of 0.32% fatal adverse
`drug reactions (ADR) (Lazarou et al., 1998) (total 6.7% ADR)
`of all hospitalized patients in the US in 1998, with a 23-fold in-
`crease of fatal ADR from 1998-2005 (Moore et al., 2007). leads
`to about 1% of hospitalized patients in the US dying from ADR.
`This suggests that drugs are not very safe, even after all the
`precautionary tests, and corresponds to the relatively frequent
`market withdrawals.
`
`The result of this disastrous situation is that pharma compa-
`nies are eating each other up, often in the hope of acquiring a
`promising drug pipeline, only to find out that this was wishful
`thinking or losing so much time in the merger that the delay of
`development compromises the launch of the pipeline drugs.
`
`Consideration 2:
`
`Clinical research, perverted by conflict
`of interest or role model?
`
`A popular criticism of clinical drug development (as, e.g..
`prominently stressed in Ben Goldacre’s recent book “Bad
`Pharma”, 2012) is the bias from the pressure to get drugs to
`the market. In fact, there is also a publication bias, i.e., the
`more successful a clinical study. the more likely it will be pub-
`lished. It has been shown that studies sponsored by industry are
`seven times more likely to have positive outcomes than those
`that are investigator-driven (Bekelman et al., 2003: Lexchin,
`2003). However, this does not take into account how much
`more development efforts go into industrial preclinical drug
`
`1 http:.f.'www.lorbas.com.'sites.fmat1hewherper.'201 2."02l1 Oithe-IruIy-staggering-cost-of-inventing—new-drugs!
`2 htlpzf/en.wikipedia.org.’wikin’List_ol_withdrawn_drugs (Accessed June 21, 2013)
`3 http:l‘.'usatoday30.usatoday.com.'news.l‘hea|lhl2009-1D-26-fda-drugs_N.htm ?csp=34
`
`276
`
`ALTEX 30, 3113
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2056 - 0002
`
`

`
`=23:
`
`HARTUNG
`
`development compared to what academic researchers have at
`their disposal.
`Actually, clinical studies have extremely high quality stand-
`ards: They are mostly randomized, double-blind, and placebo-
`controlled, as well as usually multi-centric. They require ethical
`review, follow Good Clinical Practice, and are carried out by
`skilled professionals. In recent years. the urge to publish and
`register has increased strongly. Clinical medicine also brought
`about Evidence-based Medicine (EBM). which we have several
`times praised as an objective, transparent, and conscientious
`way to condense information for a given controversial question
`(Hoffmann and l-Iartung. 2006; Hartung. 2009a, 2010). All to-
`gether, these attributes are difficult to match in other fields.
`So we might say that clinical research is pretty good even in
`acknowledging its biases, if at all. of overestimating success. ln
`a simple view, the clinical pipeline, despite enormous financial
`pressures, has very sophisticated tools to promote good science.
`If this is true, we put our money on the wrong horses in clinical
`research to begin with. We have to analyze the weaknesses of
`the preclinical phase to understand why we are not improving
`attrition rates.
`
`Consideration 3:
`
`Bushing onimol toxicology ogoin?
`
`Sure, to some extent. It is one purpose of this series of articles
`to collect arguments for transitioning to new tools. The quoted
`data from Arrowsmith would suggest that toxic side-effects
`contribute to 20% of attrition each in phase II and III. Probably,
`we need to add some percent for side-effects noted in phase I,
`i.e., first in humans, and post-market adverse reactions. Thus an
`overall figure of 30-40% seems realistic.
`However, we first have to distinguish two matters: One is the
`observed effects in humans, which were not sufficiently antici-
`pated. Another is the findings in animal toxicity studies done in
`parallel to the clinical studies. It is a common misunderstand-
`ing among lay audiences that clinical studies commence after
`toxicology has been completed. For reasons of timing. however,
`this is not possible, and the long-lasting studies are done at least
`in parallel to phase 11. Currently, when first acquiring data on
`humans. animal toxicology is incomplete. The two types of tox-
`icological data also are very different: The toxicological effects
`observed in human trials of necessarily short duration and little
`or no follow-up observation are necessarily different from the
`chronic systemic animal studies at higher doses. Fortunately,
`typical side-effects in clinical trials are mild, the most common
`one (about halfof the cases) is drug-induced liver injury (DILI),
`observed as a painless and normally easily reversible increase
`in liver enzymes in blood work though possibly extending to
`the more severe and life-threatening liver failure. The Innova-
`tive Medicine Initiative has tackled this problem in a project
`
`4 http:ttwww.imi.europaeutcontenttmip-dili
`
`based on an initiative we started with industry at ECVAM:
`“Many medicines are hurntful to the liver. and drug-incluced liv-
`er injury (DIL1) now ranks as the leading cause of liver failure
`and transplantation in western countries. However, predicting
`which drugs will prove trtxtc to the liver is extremely dtfficttlt,
`and often problems are not detected until at drug is already on
`the market.”4 The hallmark paper by Olson et al. (2000) gives us
`some idea of this and the retrospective value of animal models
`in identifying such problems: “Liver toxicity was only the fourth
`most frequent HT [human t0xictty}..., yet it led to the second
`highest terttiimttion rate. There was also less concordance be-
`tween atzimal and human toxicity with regard to liver function,
`despite liver toxicity being common in such studies. There was
`no relation between liver H73‘ and therapeutic clc.r.s'.\'."
`A completely different question is: What animal findings ob-
`tained parallel to clinical trials lead to abandoning substances?
`Probably not that many. Cancer studies are notoriously false
`positive (Basketter et al., 2012), even for almost half of the
`tested drugs on the market: furthermore. genotoxicants usu-
`ally have been excluded earlier. Reproductive toxicity will lead
`mainly to a warning against using the substance in pregnancy,
`which is a default for any new drug, as nobody dares to test
`on pregnant women. The acute and topical toxicities have been
`evaluated before being applied to humans. The same holds true
`for safety pharmacology, i.e., the assessment of cardiovascular,
`respiratory, and neurobehavioral effects, as well as excess target
`pharmacology. This leaves us with organ toxicities in chronic
`studies. In fact, if not sorted out by “investigative" toxicology,
`this can impede or delay drug development. “Fortunately,” dif-
`ferent animal species often do not agree as to the organ of toxic-
`ity manifestation, leaving open a lot of room for discussion as to
`translation to humans.
`
`Compared to clinical studies, toxicology has some advan-
`tages and some disadvantages as to quality: First, there are
`internationally harmonized protocols (especially ICI-I and
`OECD) and Good Laboratory Practice to quality-assure their
`execution. However, we use outdated methods, mainly intro-
`duced before 1970. which were systematically rendered pre-
`cautionary/oversensitive. e.g., by using extremely high doses.
`The mechanistic thinking of a modern toxicology comes as
`“mustard after the meal," mainly to argue why the findings are
`not relevant to humans. What is most evident when compar-
`ing approaches: clinical studies have one endpoint. good sta-
`tistics, and hundreds to thousands of treated individuals with
`relevant exposures. Toxicology does just the opposite: Group
`sizes of identical twins (inbred strains) are minimal, and we
`study a large array of endpoints at often “maximum tolerated
`doses" without proper statistics. The only reason is feasibility,
`but these compromises combine in the end to determine the
`relevance of the prediction made. We have made these points
`in more detail earlier (Hartung, 2008a, 200%). For a some-
`what different presentation. please see Table I which combines
`
`ALTEX 30, 30.1‘ I 3
`
`277
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2056 - 0003
`
`

`
`HARTUNG
`
`D
`
`Tab. 1: Differences between and methodological problems of animal and human studies critical to prediction
`of substance effects
`
`Subjects
`0 Small groups of (often inbred, homogenous genetic background) animals vs. large groups of individuals with heterogeneous
`genetic background
`0 Young adult animals vs. all ages in human trials
`- Animals typically only of one gender
`- Disparate animal species and strains, with a variety of metabolic pathways and drug metabolites, leading to variation in
`efficacy and toxicity
`Disease models
`
`0 Artificial diseases, i.e., different models for inducing illness in healthy animals or injury with varying similarity to the human
`condition of sick people
`0 Acute animal models for chronic phenomena
`- Monofactorial disease models vs. multifactorial ones in humans
`
`- Especially in l<nock—out mouse models the adaptive responses in animals are underestimated compensating for the knock-out
`Doses
`
`0 Variations in drug dosing schedules (therapeutic to toxic) and regimens (usually once daily) that are of uncertain relevance
`to the human condition (therapeutic optimum)
`- Pharmaco- and toxicokinetics of substances differ between animals and humans
`
`circumstances
`
`- Uniform, optimal housing and nutrition vs. variable human situations
`- Animals are stressed
`
`0 Never concomitant therapy vs. frequent ones in humans
`
`Diagnostic procedures
`- No vs. intense verbal contact
`
`I Limited vs. extensive physical exam in humans
`- Limited standardized vs. individualized clinical laboratory examination in humans
`- Predetermined timing vs. individualized one in humans
`- Extensive histopathology vs. exceptional one in humans
`0 Length of follow up before determination of disease outcome varies and may not correspond to disease latency in humans
`- Especially in toxicological studies the prevalence of health effects is rarely considered when interpreting data
`
`Study design
`- Variability in the way animals are selected for study, methods of randomization, choice of comparison therapy (none, placebo, vehicle),
`and reporting of loss to follow up
`- Small experimental groups with inadequate power, simplistic statistical analysis that does not account for potential confounding,
`and failure to follow intention to treat principles
`0 Nuances in laboratory technique that may influence results may be neither recognized nor reported. e.g.. methods for blinding
`investigators
`- Selection of a variety of outcome measures, which may be disease surrogates or precursors and which are of uncertain relevance
`to the human clinical condition
`
`- Traditional designs, especially of guideline studies, offering standardization but prohibiting progress
`
`The table combines arguments from (Olson et al., 2000; Pound et al., 2004; and Hartung, 2008a).
`
`arguments from different sources (Pound et al., 2004; Olson
`et al., 2000; Hartung, 200821) showing reasons for differences
`between animal studies and human trials.
`
`Consideration 4:
`Sorting out substances with precautionary
`toxicology before clinical studies?
`The case of genofoxicily assays
`
`Perhaps the even more important question with regard to attri-
`tion is, which substances never make it to clinical trials, that
`
`would have succeeded but whose progress was hindered by
`wrong or precautionary toxicology‘?Again we have to ask, what
`findings lead to the abandonment of a substance. This is more
`complicated than it seems, because it depends on when in the
`development process such findings are obtained and what the
`indication of the drug is. To put it simply, a new chemotherapy
`will not be affected very much by any toxicological finding.
`In early screening, we tend to be generous in excluding sub-
`stances that appear to have liabilities. An interesting case here
`is gcnotoxicity — due to the fear of contributing to cancer and
`the difficulty of identifying human carcinogens at all, this often
`is a brick wall. in addition, the relatively easy and cheap as-
`
`278
`
`ALTEX 30,3113
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2056 - 0004
`
`

`
`—t:>>
`
`HARTUNG
`
`sessment of genotoxicity with a few in vitro tests allows front-
`loading of such tests. Typically, substances will be sorted out if
`found positive. The 2005 publication of Kirkland et al. gave the
`stunning result that while the combination of three genotoxicity
`tests achieves a reasonable sensitivity of 90+% for rat carcino-
`gens, also more than 90% of non-carcinogens are false positive,
`i.e., a miserable specificity. Among the false positives are com-
`mon table salt and sugar (Pottenger et al., 2007). With such a
`high false positive rate, we would eliminate an incredibly large
`part of the chemical universe at this stage.
`This view has been largely adapted, leading to an ECVAM
`workshop (Kirkland et al., 2007) and follow-up work (Lorge et
`al., 2008; Fellows et al., 2008; Pfuhler et al., 2009, 2010; Kirk-
`land, 20l0a,b; Fowler et al., 2012a,b) financed by Cosmetics
`Europe and ECVAM, and finally changes in the International
`Conference on Harmonization (ICH) guidance, though not yet
`at the OECD, which did not go along with the suggested 10-fold
`reduction in test dose for the mammalian assays.
`However, the “false positive” genotoxicity issue (Mouse Lym-
`phoma assay and Chromosomal Aberration assay) has been chal-
`lenged more recently. Goilapucli et al. from Dow presented an
`analysis of the Mouse Lymphoma Assay at SOT 2012. “Since
`the MLA has undergone significant procedural enhancements in
`recent years, a project was undertaken to reevaluate the NTP
`data according to the current standards (IWGT) to assess the
`assay performance capabilities. Data from more than 1900 ex-
`periments representing 342 chemicals were examined against
`acceptance criteria for background mutant frequency, cloning
`efiiciency, positive control values, and appropriate dose selec-
`tion. In this reanalysis, only 17% of the experiments and 40% of
`the “positive” calls met the current acceptance standards. Ap-
`proximately 20% of the test chemicals required >1000 ug /mL
`to Satisfy the criteria for the selection of the top concentration.
`When the concentration is expressed in molarity, approximately
`58, 32, and 10% of the chemicals required 51 mM, >1 to 510
`mM, and >10 mM, respectively, to meet the criteria for the top
`concentration. More than 60% of the chemicals were judged as
`having insufiicient data to classify them as positive, negative, or
`equivocal. Of the 265 chemicalsfrom this list evaluated by Kirk-
`land et al. (2005, Mutat Res., 584, I), there was agreement be-
`tween Kirkland calls and our calls for 32% of the chemicals.”
`Astra-Zeneca (Fellows et al., 2011) published their most re-
`cent assessment of 355 drugs and found 5% unexplained posi-
`tives in the Mouse Lymphoma Assay: “Of the 355 compounds
`tested, only 52 (15%) gave positive results so, even if it is as-
`sumed that all of these are non-carcinogens, the incidence of
`‘false positive’ predictions of carcinogenicity is much lower
`than the 61% apparentfrom analysis of the literature. Further-
`more. only I 9 compounds (5%) were positive by a mechanism
`that could not be associated with the compounds primary phar-
`macological activity or positive responses in other genotoxicity
`assays.”
`Snyder and Green (2001) earlier found less dramatic false
`positive rates for marketed drugs. FDA CDER did a survey on
`
`5 http:lt'ec.europa.eulheaIth.'ph_risklcommitteesI04_socpIdocslsocp_s_08.pdf
`3 http'.I.Iwww.ebtox.com
`
`the most recent «-750 drugs and found that positive mammalian
`genotoxicity results (CA or MLA) did not affect drug approval
`substantially (Dr Rosalie Elesprue, personal communication).
`Only 1% was put on hold for this cause. However, this obvious-
`ly addresses a much later stage of drug development, at which
`most genotoxic substances already have been excluded.
`In contrast, an analysis by Dr Peter Kasper of nearly 600
`pharmaceuticals submitted to the Gennan medicines authority
`(BfArM) between 1995 and 2005, gave 25-36% positive results
`in one or more mammalian cell tests, and yet few were carcino-
`genic (Blakey et al., 2008). It is worth noting that an evaluation
`by the Scientific Committee on Consumer Products (SCCP) of
`genotoxicity/mutagenicity testing of cosmetic ingredients with-
`out animal experiments5 showed that 24 hair dyes tested posi-
`tive in vitro were all then found negative in vivo. This would be
`very much in line with the Kirkland et al. analysis. However, we
`argued earlier (Hartung, 2008b}: “The question might, however,
`be raised whether mutagenicity in human cells should be ruled
`out at all by an animal test. A genotoxic eflect in vitro shows
`that the substance has a property, which could be hazardous.
`Differences in the in vivo test can be either species-specific (rat
`versus human) or due to kinetics (does not reach the tissue at
`sufiiciently high concentrations). These do not necessarily rule
`out a hazard toward humans, especially in chronic situations or
`hypersensitive individuals. This means that the animal experi-
`ment may possibly hide a hazardfor humans.”
`In conclusion, flaws in the current genotoxicity test battery
`are obvious. There is promise of new methods, most obviously
`of the micronucleus test, which was formally validated and led
`to an OECD test guideline. There is some validation for the
`COMET assay (Ersson et al., 2013), which compared 27 sam-
`ples in 14 laboratories using their own protocols; the variance
`observed was mainly between laboratories/protocols, i.e., 79%.
`Thus standardization of the COMET assay is essential, and we
`are desperately awaiting the results of the Japanese validation
`study for the CONIET assay in vivo and in vitro. New assays
`based, e.g., on DNA repair measurement promise better accu-
`racy (e.g., Walmsley, 2008; Moreno-Villanueva et al., 2009,
`201 1). Whether the current data justify eliminating the standard
`in vitro tests and adopting the in vivo comet assay as specified in
`the new ICI-I S2 guidance before validation can be debated. This
`guidance in fact decreases in vitro testing and increases in viva
`testing (in its option 2 as it replaces in vitro mammalian tests
`entirely with two in vivo tests). It is claimed that they can be
`done within ongoing sub-chronic testing, but this still needs to
`be shown because the animal genotoxicity tests require a short
`term (2-3 day) high dose, while the sub-chronic testing neces-
`sitates lower doses.
`
`What to do‘? We need an objective assessment of the evidence
`concerning the reality of “false positives.” This could be a very
`promising topic for an evidence—based toxicology collaboration
`(EBTC") working group. Better still, we should try to find a bet-
`ter way to assess human cancer risk without animal testing. The
`animal tests are not sufficiently informative.
`
`ALTEX 30. 30/13
`
`279
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2056 - 0005
`
`

`
`HARTUNG
`
`lgj
`
`What does this mean in the context of the discussion here?
`
`lt shows that even the most advanced use of in vitro assays to
`guide drug development is not really satisfactory. Though the
`extent of false positives, i.e., innocent substances not likely to
`be developed further to become drugs. is under debate. it ap-
`pears that no definitive tool for such decisions is available. The
`respective animal experiment does not offer a solution to the
`problem, as it appears to lack sensitivity. Thus, the question
`remains whether genotoxicity as currently applied guides our
`drug development well enough.
`
`Consideration 5:
`
`If animals were fortune tellers of drug efiicacy,
`they would not make a lot of money...
`
`A large part of biomedical research relies on animals. John Io-
`annidis recently showed that almost a quarter of the articles in
`PubMed show up with the search term “animal .” even a little
`more than with “patient" (loannidis, 2012). While there is in-
`creasing acknowledgement that animal tests have severe limi-
`tations for toxicity assessments, we do not see the same level
`of awareness for disease models. The hype about genetically
`modified animal models has fueled this naive appreciation of
`the value of animal models.
`
`The author had the privilege to serve on the National Academy
`of Science panel on animal models for countenrieasures to bio-
`terrorism. We have discussed this recently (Hartung and Zurlo,
`2012): The problem for developing and stockpiling drugs for the
`event of biologicallchemical terrorism or warfare is that (fortu-
`nately) there are no patients to test on. So, the question to the
`panel was how to substitute in line with the animal rule of FDA
`with suitable animal models. in a nutshell, our answer is: There
`are no such things as sufficiently predictive animal models to sub-
`stitute for clinical trials (NRC. 2011). Any drug company would
`long to have such models for drug development, as the bulk of
`development costs is incurred in the clinical phase; for counter-
`measures we have the even more difficult situation of unknown
`
`pathophysiology, limitations to experiment in biosafety facilities,
`disease agents potentially designed to resist interventions, and
`mostly peracute diseases to start with. So an important part of the
`committee’s discussions dealt with the attrition (failure) rate of
`drugs entering clinical trials (see above). which does not encour-
`age using animal models to substitute for clinical trials at all.
`In line with this, a recent paper by Seok et al. (2013) showed
`the lack of correspondence of mouse and human responses in
`sepsis, probably the clinical condition closest to biological war-
`fare and terrorism. We discussed this earlier (Leist and Hartung,
`2013) and here only one point shall be repeated, i.e., though not
`necessarily as prominent and extensive. several assessments of
`animal models led to disappointing results, as referenced in the
`comment for stroke research.
`
`In toxicology. we have seen that different laboratory species
`exposed to the same high doses predict each other no better than
`
`7 htlprllwww.nc3rs.org.ukldown|oaddoc.asp?icl=695
`3 http:llwww.wc8.ccac.calfiIest'WC6_DecIaration_ol_Montrea|_FlNAL.pdl
`
`60% -— and there is no reason to assume that any of them predict
`humans better at low doses. We lack such analysis for drug ef-
`ficacy models systematically comparing outcomes in different
`strains or species of laboratory animals. It is unlikely that results
`are much better.
`
`In this series ( Hartung , 20083) We have addressed the shortcom-
`ings of animal tests in general terms. Since then, the weaknesses
`in quality and reporting of animal studies, especially. have been
`demonstrated (MacCallum, 2010; Macleod and van der Worp.
`2010; Kilkenny et al., 2010; van der Worp and Macleod, 2011),
`further undermining their value. Randomization and blinding
`rarely are reported, which can have important implications. as
`it has been shown that animal experiments carried out without
`either are five times more likely to report a positive treatment
`effect (Bebarta et al., 2003). Baker et al. (2012) recently gave an
`illustration of poor reporting on animal experiments, stating that
`in "180 papers on multiple sclerosis listed on PubMed in the past
`6 months, we found that only 40% used appropriate .s'tatistic'.\‘ to
`compare the efifects of gene-knockozit or treatment. Appropriate
`statistics were applied in only 4% of neuroimrnttnological stud-
`ies published in the past two years in Nature Pirlalishing Group
`journals, Science and Cell” (Baker et al., 2012).
`Some more systematic reviews of the predictive value of
`animal models have been little favorable, see Table 2 (Roberts,
`2002: Pound et al., 2004; Hackam and Redelmeier, 2006: Perel
`
`et al., 2007: Hackam, 2007: van der Worp et al., 2010). Hack-
`man and Rede1meier(Hackam and Redelmeier, 2006). for exam-
`ple, found that of 76 highly cited animal studies, 28 (37%: 95%
`confidence interval [CI]. 26%-48%) were replicated in human
`randomized trials. 14 (18%) were contradicted by randomized
`trials,and 34 (45%) remain untested. This is actually not too bad,
`but the bias to highly cited studies (range 639 to 2233) already
`indicates that these studies survived later repetitions and transla-
`tion to humans. There are now even more or less "systematic"
`reviews of the systematic reviews (Pound et al.. 2004: Mignini
`and Khan, 2006: Knight, 2007: Briel et al., 2013). showing that
`there is room for improvement. They definitely do not have the
`standard of evidence-based medicine. In the context of evidence-
`
`based med icine, “A systematic review involves the application of
`scientific strategies, in ways that limit bias, to the assembly, crit-
`ical appraisal, and .s'yn.thesis of all relevrmt sturiies that (address
`a specific clinical question” (Cook et al.. 1997). But the concept
`is maturing. See. for example. the NC3R whitepaper “S_vstem.atic'
`reviews of animal research"7 or the “Montréal Declaration on
`Systematic Reviews of Animal Stuclies."8 The ARRIVE guide-
`line (Kilkenny et al., 2010) and the Gold Standard Publication
`Checklist (GSPC) to improve the quality of animal studies (Hoo-
`ijmans et al., 2010) facilitate the evaluation and standardization
`of publications on animal studies.
`No wonder that in vitro studies are increasingly considered:
`“According to a new market report by Transparency Market
`Research. the global in vitro toxicity tes

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