banner



Why Do We Use Animals In Toxicological Studies, Instead Of Humans?

  • Journal List
  • JACC Bones Transl Sci
  • 5.4(seven); 2019 Nov
  • PMC6978558

JACC Bones Transl Sci. 2019 November; 4(7): 845–854.

Limitations of Animal Studies for Predicting Toxicity in Clinical Trials

Is information technology Time to Rethink Our Electric current Approach?

Received 2019 Oct xiv; Accustomed 2019 Oct 14.

Summary

Brute testing is used in pharmaceutical and industrial research to predict human toxicity, and nevertheless analysis suggests that beast models are poor predictors of drug safety in humans. The cost of creature inquiry is loftier—in dollars, delays in drug approval, and in the loss of potentially beneficial drugs for human apply. Human subjects accept been harmed in the clinical testing of drugs that were accounted rubber past animal studies. Increasingly, investigators are questioning the scientific merit of animal research. This review discusses issues in using animals to predict human toxicity in pharmaceutical evolution. Role one focuses on scientific concerns over the validity of creature enquiry. Part two will discuss alternatives to animal inquiry and their validation and use in production of human pharmaceuticals.

Key Words: animate being research, drug development, toxicity, translational inquiry

Abbreviations and Acronyms: FDA, U.Due south. Nutrient and Drug Assistants; LR, likelihood ratio; NLR, negative likelihood ratio; NPV, negative predictive value; PLR, positive likelihood ratio; PPV, positive predictive value

Graphical abstract

An external file that holds a picture, illustration, etc.  Object name is fx1.jpg

In that location is no dubiousness that the use of animals in science and medicine has significantly benefitted homo beings (Tabular array 1). However, many investigators are increasingly concerned that animal experimentation may be based on a scientifically flawed premise and that it retains its acceptability only because clear alternatives take not been identified. Dramatically rising costs and extremely high failure rates in drug evolution have led many to re-evaluate the value of animal studies. This review focuses on questions regarding the scientific validity of nonhuman animate being models (hereafter referred to but every bit "animal enquiry") in predicting homo toxicity in preclinical pharmaceutical testing.

Table i

Fauna Uses in Science, Medicine, and Research

Predictive models for human being diseases and their processes
Predictive models for testing drugs and other chemicals for man toxicity and efficacy
"Spare parts"— eastward.g., pig-derived aortic valve prostheses
Bioreactors or factories— east.g., production of monoclonal antibodies
Sources of tissue to report physiological principles
Educational "material" to educate and train biology and medical students and others
Subjects in research to do good other animals
Subjects of research to gain basic knowledge for its own sake

Historical Perspective

In the United States, the use of animals to examination human pharmaceuticals dates to 1937, when a liquid formulation of a sulfa antibiotic dissolved in ethylene glycol resulted in the deaths of 107 adults and children. The incident resulted in passage of the 1938 U.S. Federal Nutrient, Drug, and Cosmetic Act, mandating animal toxicity testing (1,ii). In 1946, linguistic communication was incorporated into the Nuremberg code (three) and after the Helsinki Proclamation (4) requiring homo experiments to be "designed and based on the results of creature experimentation [writer's italics] and a knowledge of the natural history of the disease." The statement was written by Andrew Ivy, a potent proponent of animal inquiry, but was not based on scientific show that such a requirement would improve rubber or efficacy of human drug evolution (3).

Today, the U.Due south. Food and Drug Administration (FDA) by and large requires preclinical testing of any new drug or biological therapeutic "for pharmacologic activeness and acute toxicity in animals" prior to inbound human being clinical trials (5). In certain cases, such every bit emergency treatment for hazardous exposure, the FDA may even corroborate in-homo use based solely on animal testing under "The Brute Efficacy Rule" (6).

Despite the securely rooted supposition that creature models accurately predict man toxicity (vii, 8, 9), fifty-fifty cursory examination of the concordance of creature and human being trials raises concerns. A 2006 review of 76 animal studies, for case, plant that approximately 20% were contradicted in humans and only 37% were ever replicated in humans (ten). A review of 221 brute experiments found agreement in human studies just fifty% of the time—substantially randomly (11). Review of 37 chemicals studied in the U.S. National Toxicology Program concluded that toxicities other than carcinogenesis were non reproducible between rats and mice, between sexes, or compared with historic control animals. Average positive predictive value (PPV) from mouse to rat was 55.3% and 44.8% for long-term and short-term studies, respectively. Combining organ, length of exposure, and sex, PPV between mice and rats hovered around l%, which is no greater than random risk (12). An analysis of 2,366 drugs concluded that "results from tests on animals (specifically rat, mouse and rabbit models) are highly inconsistent predictors of toxic responses in humans, and are little meliorate than what would result merely past gamble—or tossing a coin—in providing a basis to decide whether a compound should proceed to testing in humans" (13). Similar results were found for nonhuman primates and dogs (fourteen). Indeed, we need get no further than the failure rates in drug development to accept serious questions about whether animal testing accurately predicts toxicity in human trials.

About 12% of pharmaceuticals pass preclinical testing to enter clinical trials (15). Of those, only 60% successfully complete phase I trials (16). Overall, approximately 89% of novel drugs fail human clinical trials, with approximately one-half of those failures due to unanticipated human toxicity (Figure 1) (17). If animal tests accurately predict human toxicity, then why are toxicity-related failure rates in human clinical trials then high?

An external file that holds a picture, illustration, etc.  Object name is gr1.jpg

Failures in Translational Enquiry: Preclinical and Clinical Trials

Percentages of drugs that neglect in preclinical trials (due to drug toxicity or failure of efficacy in fauna testing) and in clinical trials (due drug toxicity or failure of efficacy in human testing) are shown in columns ane and 2. The tertiary column demonstrates what would happen if animal and man toxicity were closely correlated and therefore drugs with man toxicity were eliminated at the preclinical testing stage by brute toxicity testing (one-half of all drug failures in clinical trials are due to toxicity issues despite safety in animals). Success rates of clinical trials increase from 11.seven% overall to approximately 56%.

The Price of Wrong Decisions

Two critical "wrong" decisions regarding animate being tests of human being pharmaceuticals are ane) falsely identifying a toxic drug as "safe" and 2) falsely labeling a potentially useful therapeutic amanuensis as toxic.

When a human being-toxic drug is identified equally "safe" past animal testing, the nearly likely effect by far is that the drug will fail in clinical testing, frequently due to unacceptable adverse human being effects, and sometimes significantly harming volunteer research subjects in the process. Drugs that survive clinical trials and reach market approval may still be recalled later due to toxicity identified just after months or years of in-human use. Vioxx (Merck, Kenilworth, New Jersey) was plant after release to significantly increase the risk of cardiovascular morbidity and mortality, costing Merck more than $viii.v billion in legal settlements solitary (18). An estimated 88,000 people suffered eye attacks later on taking Vioxx and 38,000 died (19).

Of 578 discontinued and withdrawn drugs in Europe and the The states, almost one-half were withdrawn or discontinued in post-approval actions due toxicity (20). Van Meer et al. (21) found that of 93 post-marketing serious adverse outcomes, just 19% were identified in preclinical animal studies. In the get-go decade of the 21st century, approximately one-third of FDA-approved drugs were after cited for safety or toxicity issues. or a combination of both, including human cardiovascular toxicity and brain harm, afterwards remaining on the market for a median of 4.2 years (22,23). The well-nigh common toxicity types associated with drug withdrawals in the The states and Europe are hepatic (21%), cardiovascular (sixteen%), hematological (11%), neurological (9%), and carcinogenicity (8%) (Figure 2) (xx).

An external file that holds a picture, illustration, etc.  Object name is gr2.jpg

Toxicity Failures in Pharmaceutical Development

Poly peptide-based biologics (e.g., monoclonal antibodies), fusion proteins, and recombinant proteins now account for nearly development stage and marketed biopharmaceuticals (15). These nowadays a particular challenge in predicting human toxicity, due to their propensity to provoke production of antidrug antibodies. Safety concerns include cross reactivity, potentially exaggerated pharmacology, and tedious recovery from toxicity, among others (15,24)—and immunogenic responses in animals do non predict immunogenicity in humans (15,25, 26, 27).

There are many notable examples of cases in which beast trials did not predict severe man toxicity. Isuprel for treatment of asthma caused over 3,500 deaths in U.k. solitary, despite condom in rats, guinea pigs, dog, and monkeys, all of which had received doses far exceeding those administered in humans (ii,28). Thalidomide acquired devastating phocomelia in an estimated 20,000 to 30,000 infants before it was withdrawn. However, creature tests failed to reveal meaning teratogenicity in 10 strains of rats; 11 breeds of rabbit; 2 breeds of domestic dog; iii strains of hamsters; viii species of primates; and various cats, armadillos, republic of guinea pigs, swine, and ferrets (29). An antibody to care for human autoimmune disease, TGN1412, was given at one/500th the dose constitute safe in animate being testing to 6 human volunteers in a stage I trial (30,31), rendering them all critically sick inside minutes and leaving them all with long-term complications (32, 33, 34). BIA-102474-101, a drug developed for a range of disorders from anxiety to Parkinsonism, caused deep brain hemorrhage and necrosis in all 5 human volunteers during a phase I clinical trial after it was administered in doses that were i/500th of the safe dose for dogs. One volunteer died (35). Fialuridine, for treatment of hepatitis B, acquired the deaths of five volunteers during stage II clinical trials despite being safe in mice, rats, dogs, monkeys, and woodchucks in doses that were hundreds of times college. Ii other volunteers only survived after receiving liver transplants (32).

When animal tests falsely identify a safe chemical every bit "toxic," the nigh sure outcome is abandonment of further evolution. Undoubtedly many potentially benign drugs have failed animal testing and been lost to patients, even though they would have been both safe and effective (36,37). Because a drug that shows toxicity in brute models is unlikely to ever undergo human being testing, the magnitude of this type of "error" is unknown. Still, many highly beneficial drugs would have failed animal testing and would never have been brought to market, except that they were developed before animate being testing was required (38). Examples include penicillin (fatal to guinea pigs) (39), paracetamol (toxic in dogs and cats) (40), and aspirin (embryo toxicity in rats and rhesus monkeys) (41).

Lack of animate being tests has also caused deleterious delays in critical drug approvals. Compassionate human employ of ganciclovir demonstrated efficacy and safe in treating acquired immunodeficiency syndrome–related cytomegalovirus retinitis in more human being patients than would generally be required for a stage I clinical trial, just the FDA refused to license it due to lack of beast studies. Ganciclovir had too been used safely in over 300 patients nether compassionate use to care for cytomegalovirus colitis—more would mostly be required in a phase II clinical trial—but the FDA delayed clinical trials for more than a twelvemonth due to lack of animal studies. The drug was finally canonical after a 4-twelvemonth delay (42).

Time and dollars

Rodent testing in cancer therapeutics adds an estimated 4 to 5 years to drug development and costs $two to $four million. For industrial toxicity testing, information technology takes virtually 10 years and $three 1000000 to complete all required animal studies to annals a unmarried pesticide (43). Compared with the costs of in vitro testing, beast tests range from 1.5× to >30× as expensive (44,45).

No comprehensive reviews of the total overall cost of animal testing in pharmaceutical evolution appear to be. In part, this may exist because fifty-fifty the total number of animals or of such studies is unknown. The 2002 amendments to the Animal Welfare Act exempted mice, rats, fish, and birds used in animal research from required reporting to the U.S. Department of Agriculture (46). These are the iv nigh mutual types of animals used, and they account for >90% of all U.S. animal subjects and 81% of European animal subjects (45,47).

Costs of animal toxicity tests can be estimated from other industries, however, and are eye-opening. According to the Organisation for Economical Development, which determines animal testing guidelines and methodology for government, industry, and contained laboratories in its several dozen member countries, the average cost of a single, ii-generation reproductive brute toxicity study worldwide is €318,295 and for Europe lone is €285,842 (45), or roughly $349,890 and $314,215, respectively.

Contract inquiry organizations account for almost of the animal testing done in the The states and Europe. Statista, a global information portal for market and economic sector statistics, estimates the global markets for animal testing in 2018 at $7.4 billion for drug discovery, $11.2 billion for preclinical development and safe, $58.v billion for clinical development, and $ii.3 billion for primal laboratory testing (48). Keen (49) estimates that annual U.S. biomedical and agricultural research and development investments involving animal enquiry exceed $26 billion.

Reproducibility and Interspecies Reliability of Animal Tests

Reproducibility of animal studies within species, fifty-fifty when carried out nether rigorous protocols, is questionable. Using a database of more than than 800,000 animal toxicity studies performed for 350 chemicals under rigorous guidelines, a reviewer found toxicity was repeatable merely 70% of the fourth dimension in the same species (45). Another reviewer found that results for a single chemical often differed with animal model, strain, dose, and delivery route. About 26% of chemicals demonstrated contradictory results on repeat testing in the same species. Furthermore, discordant results sometimes ranged over 3 orders of magnitude within the same species (50).

PPV, NPV, and LR

Sensitivity reflects how likely a positive test is to detect all subjects with a condition, and specificity reflects how likely a negative test is to exclude all subjects without the condition (Figure iii). PPV reflects how oft a positive test actually identifies a subject with the condition, and the power of the negative predictive value (NPV) of a test reflects the proportion of subjects with negative tests that really do not accept the status. Whereas sensitivity, specificity, PPV, and NPV are ofttimes used to describe the accuracy of tests, they are non sufficient to inform usa how much "value" to attribute to whatever given examination. For case, suppose a positive toxicity test in mice for a group of drugs ever predicts human being toxicity (sensitivity = 100%), just it too indicates human being toxicity when it is not present—in fact the examination results always indicate that the drug is toxic. Such a test would have almost no use in determining human toxicity despite being 100% sensitive. A useful toxicity exam is 1 that also indicates accurately when toxicity in animals is not nowadays in humans or has high specificity. Furthermore, we want to know how ofttimes the test accurately indicates man toxicity, compared with how accurately it indicates human being nontoxicity.

An external file that holds a picture, illustration, etc.  Object name is gr3.jpg

Computing LR

The likelihood that a test showing toxicity in a mouse predicts toxicity in the rat (positive likelihood ratio [PLR]) or that a test showing no toxicity in a mouse predicts nontoxicity in a rat (negative likelihood ratio [NLR]). M+R+ = toxicity nowadays in both mouse and rat; M+R = toxicity present in mouse but non in rat; ThouR+ = toxicity not present in mouse, just present in rat; and MR = toxicity not nowadays in mouse and likewise not present in rat.

Sensitivity, specificity, PPV, and NPV are all strongly affected by the prevalence of the condition they test for and are therefore of limited value in assessing the reliability of a test when the prevalence of the condition is unknown. Lower prevalence increases the likelihood of fake positive results, and higher prevalence increases the likelihood of false negative results. Once a drug tests positive for toxicity in animals, information technology is rarely tested against humans, and the prevalence of the real status the exam is being used to "detect"—human toxicity—remains unknown.

Nonetheless, the "value" of using a given test to ameliorate the post-test probability of ruling in or ruling out a condition can be calculated using likelihood ratios even if the prevalence of the condition is unknown, so long as the sensitivity and specificity of the test are known. LR are indicators of whether the results of a given test will "add weight" over the pre-examination probabilities (i.eastward., prevalence rate) of the condition in deciding what the probability is that a condition is actually present or absent.

At that place are 2 types of LR: the positive likelihood ratio (PLR) indicates how much more likely it is that a condition exists later on a positive examination result, when compared with its pre-exam probability. The negative likelihood ratio (NLR) indicates how much the probability that a condition exists decreases compared with its pre-test probability, given a negative examination result. The modify in post-test probability from pre-test probability is calculated past multiplying the pre-test probability (prevalence) by the PLR or NLR. If the change in post-test probability from the pre-test probability is minor (i.e., LR, the multiplier, is small), then the test is unlikely to help determine the presence or absenteeism of a condition over but knowing its prevalence. LR of <ane.0 actually indicate a negative shift in post-exam probabilities. In other words, if a PLR is <1.0, and then for whatever field of study that has a positive test result, the probability that they have the condition decreases compared with the pre-exam probability. For an NLR of <one.0, for any subject with a negative exam, the probability that they do not have the condition also decreases compared with the pre-test probability. For an LR of 1.0, there is no change from pre-test probabilities (pre-test probabilities are simply multiplied by 1), and the test also was not useful. For LR >ane.0, the probability of the condition being nowadays increases in the face of a positive examination, and the probability of the condition being absent increases in the presence of a negative test. For LR from i.0 to 10, these changes are relatively small-scale (meaning the exam will not add much), but for LR >ten, the changes increase exponentially and are considered meaning (51, 52, 53, 54).

Using LR to calculate the probability that a test volition improve detection of a condition or ruling it out is complex; information technology requires knowing the sensitivity and specificity of a examination and pre-examination probabilities, conversion of probabilities to odds and back again, and then using a log tabular array (i.eastward., a Fagan'due south nomogram) or log calculator to determine how much a test is likely to better (or subtract) the chances of detecting the condition (53).

LR are increasingly being used to express translatability of animal toxicity testing (52, 53, 54, 55). Bailey et al. (14) establish that the presence of toxicity in a species sometimes added evidentiary weight to the run a risk of toxicity in another, only the reverse was not true: negative toxicity tests in animals did non significantly increase the probability that a toxic exam would also be negative in humans, and a lack of toxicity in any species would not reliably point a likely lack of toxicity in any other species, including comparisons of primate to human toxicity tests (xiv). Furthermore, even in the presence of brute toxicity, LRs were extremely inconsistent and varied considerably for different classes of drugs (thirteen,52). Like findings have been reported in multiple analyses and reviews in other studies (52,54, 55, 56).

A number of studies have reviewed LR of specific drug toxicity tests for which both brute and human data are available. In a review of 2,366 drugs, including data from 3 of the most common animal enquiry species—rat, mouse, and rabbit—PLRs were by and large high (i.e., in that location is a likelihood that positive toxicity tests in animals would show toxicity in humans). But median NLRs were very low—1.12 (rabbit), i.39 (mouse), and 1.82 (rat); in other words, they were of little or no value in excluding man toxicity (13). The investigators too examined canine models and found that PPV and PLR for human toxicity were not correlated with 1 another: NLR were low, indicating that the dog provided little evidentiary weight to ruling out toxicity in humans (52). Afterwards analysis of 3,000 drugs establish that tests inferring no toxicity in whatever 1 species, including nonhuman primates, have no evidentiary weight with regard to toxicity in whatsoever other species (xiv). In a comparison study reported by pharmaceutical companies of 150 drugs associated with adverse events or toxicity in humans (55), LR could not exist determined due to a lack of specificity reporting on the tests. Paglialunga et al. (56) examined translatability of respiratory safety pharmacology studies from animate being models to humans and found that PPV and PLR were so low that animal tests provided little value in predicting human being toxicity.

Growing Scientific Criticism

Equally early every bit 1962, scientists questioned the supposition that brute models reliably predicted human responses. Lichtfield (57) examined 6 drugs studied in animate being models and establish that rats and dogs demonstrated PPVs (for human response) of 0.49 and 0.55, respectively, essentially random hazard. He opined that the differences between species in specific drug responses were so hitting that one could actually use the results of drug toxicity tests alone to identify whether an entity was a rat, rather than a dog or a human, and concluded in that location was no ground for predicting adverse human furnishings for the 6 drugs from beast studies. A 1990 assay of the toxicities of 24 drugs abandoned during human clinical trials demonstrated that 16 had no creature model toxicity correlation (58).

In 1981, the Council on Scientific Affairs of the American Medical Association stated, "The Council's consultants hold that to identify carcinogenicity in brute tests does non per se predict either risk or event in human experience. . . . the Council is concerned nearly the hundred[southward] of millions of dollars that are spent each year (both in the public and individual sectors) for the carcinogenicity testing of chemical substances. The concern is particularly grave in view of the questionable scientific value of the tests when used to predict the man experience" (59).

The 2019 West Coast Regional Safety Pharmacology Society Meeting discussed concerns nearly the lack of concordance between animal and homo safe studies, including lack of canine and man concordance for proarrhythmia risks of new cardiovascular drugs and the failure of animate being research to predict drug-related risks in the human fundamental nervous and respiratory systems (60,61).

Regulatory and research leaders are increasingly taking notice of the issue. In 2006, Michael Levitt, and then U.South. Secretary of Health and Homo Services, stated, "nine out of ten experimental drugs fail in clinical studies because we cannot accurately predict how they will behave in people based on laboratory and animate being studies" (62,63). A landmark review and report by the Institute of Medicine in 2011 concluded that the employ of chimpanzees in biomedical research is unnecessary (64). Although the reasons for information technology are complex, in 2015, the National Institutes of Health announced they would be ending all chimpanzee research (65). Andrew Wheeler, ambassador of the U.Southward. Ecology Protection Bureau, pledged in September 2019 to phase out all toxicity testing in mammals over the next 16 years (66).

Is the Scientific Premise Behind Beast Models Valid?

Many concerns regarding reliability of animal models in predicting human toxicity are not based on the scientific underpinnings of interspecies translation, but rather call out collateral, potentially correctable issues, such as technical competence in executing beast enquiry, the soundness of animal enquiry report design, and publication bias (Table 2) (67, 68, 69, 70, 71). Indeed, Knight (72) could observe no review of animal research studies that rated a bulk of the experiments as having "good" methodological quality. An obvious solution would be correction of these problems to ameliorate translation rates of beast research. Despite widespread efforts to improve the quality of methodology in animal studies, however, studies examining whether such measures consistently ameliorate the reliability of animal models in predicting man toxicity have yet to be published, although a number of studies practice demonstrate continued issues with predicting human efficacy (73).

Table 2

Ordinarily Used Arguments Confronting Animal Inquiry

Argument Critique
Methodological: Animal models should be abandoned because the scientific methodology of the experiment was poor. The quality of methodology in an individual experiment cannot exist extrapolated to the question of whether animate being experimentation as a whole is invalid, simply to whether the private experiment is yielding true results.
Historical: Historically, medical dependence on fauna modeling is much less robust than we are led to believe. Historical use of animal modeling is a poor measure out of the validity of current experimentation and methods. To determine whether fauna modeling is reliable in current scientific discipline, we need to use mod scientific knowledge and examine modern methodology to determine whether animal modeling is predictive of human outcomes today. This takes into account information and methods that may or may non have been historically available.
Reviews: Review manufactures have determined that certain animal species have non been disquisitional in various medical developments, and therefore animal experimentation should exist abolished. The invalidity of using certain specific animals does not necessarily rule out animal models every bit a whole.
Alternatives: The beingness of alternative models requires u.s. to carelessness animal research. Whereas alternatives to fauna research exist or are developing in many areas of medical enquiry, in many instances such alternatives do not be. This argument does not address whether continued utilise of animal models is scientifically valid, regardless of alternative methods, and it does not attempt to ascertain whether certain animal models are predictably successful and others are predictably unsuccessful.

Instead of methodologies and publication bias, an increasing number of investigators propose that the problem may prevarication with the basic premise of creature testing itself (69,74). The biological sciences take increasingly embraced theories regarding complex systems (e.g., anarchy theory and complexity theory) to explain mechanisms in evolution, the biology of cancer, the divergent backdrop of animal species, every bit well as the failures of translation of drug therapeutics from animal species to humans (75,76). Because animals and humans are classic examples of incompletely understood complex systems, some investigators propose that it may simply exist scientifically invalid to assume that toxicity of a substance in any one species can reliably predict toxicity in any other, no matter how stringent animal testing standards are fabricated (69).

Alternatives to Animal Testing

Alternatives to animal testing will be discussed in more than detail in office 2 of this review; they include in vitro tests using prison cell lines, tissue samples, use of culling organisms such equally bacteria, iii-dimensional modeling and bioprinting, in silico tests, organ-on-scrap technologies such as 3-dimensional organoids, computer modeling, and phase 0 in-human being microdosing trials (77, 78, 79, 80, 81, 82). A comprehensive report of the accuracy, LR, and costs of alternative testing methods compared with creature toxicity testing has not been published; even so, at that place is data suggesting that in vitro testing and other methods are significantly faster and less expensive than animal models (42,44). Using homo cells, tissue, or organ models to form the footing of an in vitro test may meliorate accurateness in weeding out drugs with meaning agin man effects; withal, this assumption, too, will require rigorous study.

Researchers will undoubtedly exist challenged sooner rather than later to reduce creature research every bit the result of public advancement efforts. A 2019 spending pecker passed by the U.Due south. House of Representatives includes a directive to the National Institutes of Wellness to advance the replacement of nonhuman primates in inquiry with alternative research models (83).

The FDA states that for the purposes of corrective testing, they believe "that prior to utilise of animals, consideration should be given to the use of scientifically valid alternative methods to whole-animate being testing" (84). The Interagency Analogous Commission on Validation of Alternative Methods and the National Toxicology Programme Interagency Center for the Evaluation of Alternative Toxicological Methods were established in 1997 to coordinate the development, validation, acceptance, and harmonization of culling toxicological test methods throughout the U.Southward. government (85) and take equally a part of their mission the explicit mandate to reduce or eliminate whole fauna testing. The Biennial Progress Written report of the Interagency Coordinating Commission on Validation of Alternative Methods for 2016 to 2017 details actions they have taken, including, amidst others: 1) publication of guidance documents waiving all astute dermal lethality studies for pesticides and describing a process for evaluating; 2) publication of notices reducing the number of hamsters for potency testing of certain vaccines; and 3) publication of a roadmap for integrating predictive toxicology methods into safe and risk assessments by the FDA (86). At this time, the FDA mostly still requires submission of preclinical brute information in investigational new drug applications (5).

Conclusions

Although creature toxicity testing has been the stalwart basis of "ensuring" safety of in-human being clinical testing and apply, examination of the published data raises significant questions about whether it is reliable and should be abandoned or at least significantly concise in favor of other potentially more reliable methods. Savings in time and cost for new therapeutics could exist substantial, if the safety of nonanimal preclinical testing is proven. Increasingly, scientific organizations and authorities regulatory agencies are recognizing that culling methods may replace animal testing and meliorate the flow and safety of new therapeutics to human use.

Footnotes

Dr. Van Norman has received fiscal back up from the Journal of the American College of Cardiology.

The writer attests they are in compliance with human studies committees and animal welfare regulations of the authors' institutions and Food and Drug Administration guidelines, including patient consent where appropriate. For more than information, visit the JACC: Basic to Translational Scienceauthor instructions folio.

References

one. Wax P.Chiliad. Elixirs, diluents and the passage of the 1938 Federal Food, Drug and Cosmetics Act. Ann Intern Med. 1995;122:456–461. [PubMed] [Google Scholar]

two. Greek R., Pippus A., Hansen Fifty.A. The Nuremberg Code subverts human wellness and safety by requiring beast modeling. BMC Med Ethics. 2012;13:xvi. [PMC free article] [PubMed] [Google Scholar]

3. National Institutes of Health . U.S. Government Printing Role; Washington, DC: 1949. Regulations and Upstanding Guidelines. Reprint from: Trials of State of war Criminals before the Nuremberg War machine Tribunals under Control Council Constabulary no. 10, Vol. 2; pp. 181–182.https://history.nih.gov/research/downloads/nuremberg.pdf Bachelor at: [Google Scholar]

7. Fomchenko Eastward.K., The netherlands E.C. Mouse models of encephalon tumors and their applications in preclinical trials. Clin Cancer Res. 2006;12:5288–5297. [PubMed] [Google Scholar]

eight. Huff J., Jacobson M.F., Davis D.L. The limits of two-year bioassay exposure regimens for identifying chemical carcinogens. Environ Health Perspect. 2008;116:1439–1442. [PMC gratis article] [PubMed] [Google Scholar]

9. Gad Due south. Preface. In: Gad S., editor. Brute Models in Toxicology. CRC Press; Boca Raton, FL: 2007. pp. i–eighteen. [Google Scholar]

10. Hackam D.M., Redelmeier D.A. Translation of research evidence from animals to humans. JAMA. 2006;296:1731–1732. [PubMed] [Google Scholar]

11. Perel P., Roberts I., Sena East. Comparing of handling effects between creature experiments and clinical trials: systematic review. BMJ. 2007;334:197–203. [PMC free commodity] [PubMed] [Google Scholar]

12. Wang B., Gray G. Cyclopedia of noncarcinogenic endpoints in rodent chemical bioassays. Risk Anal. 2015;35:1154–1166. [PubMed] [Google Scholar]

thirteen. Bailey J., Thew M., Balls Yard. An assay of the use of animal models in predicting human toxicology and drug condom. Altern Lab Anim. 2014;42:189–199. [PubMed] [Google Scholar]

14. Bailey J., Thew M., Balls M. Predicting human drug toxicity and safety via brute tests: tin any ane species predict drug toxicity in any other, and do monkeys help? Altern Lab Anim. 2015;43:393–403. [PubMed] [Google Scholar]

16. Van Norman G.A. Drugs, devices and the FDA: function 1: an overview of approving processes for drugs. J Am Coll Cardiol Basic Trans Science. 2016;1:170–179. [PMC free article] [PubMed] [Google Scholar]

17. Van Norman G.A. Phase Two trials in drug development and adaptive trial design. J Am Coll Cardiol Bones Trans Science. 2019;iv:428–437. [PMC free article] [PubMed] [Google Scholar]

19. Juni P., Dip L.N., Reichenback South., Sterchi R., Dieppe P.A., Egger M. Risk of cardiovascular events and rofecoxib: cumulative meta-analysis. Lancet. 2004;364:2021–2029. [PubMed] [Google Scholar]

xx. Siramshetty B.V., Nickel J., Omieczynski C. WITHDRAWN—a resources for withdrawn and discontinued drugs. Nucl Acids Res. 2016;44:D1080–D1086. [PMC gratis article] [PubMed] [Google Scholar]

21. van Meer P.J., Kooijman One thousand., Gispen-de Wied C.C., Moors Due east.H., Schellekens H. The ability of animal studies to detect serious postal service marketing agin events is express. Reg Tox Pharm. 2012;64:345–349. [PubMed] [Google Scholar]

23. Downing N.Southward., Shah Due north.D., Aminawung J.A. Postmarket safe events among novel therapeutics approved past the United states of america Food and Drug Administration between 2001 and 2010. JAMA. 2017;317:1854–1863. [PMC costless commodity] [PubMed] [Google Scholar]

24. Brinks V., Jiskoot W., Schellekns H. Immunogenicity of therapeutic proteins: the use of animal models. Pharm Res. 2011;28:2379–2385. [PMC free article] [PubMed] [Google Scholar]

26. De Groot A.Due south., Moise L. Prediction of immunogenicity for therapeutic proteins: country of the fine art. Curr Opin Drug Discov Devel. 2007;10:332–340. [PubMed] [Google Scholar]

27. Pedras-Vasconcelos J.A. The Immunogenicity of therapeutic Proteins—What You Don't Know Tin can Injure You lot and the Patient. U.S. Food and Drug Administration. Fall 2014. https://world wide web.fda.gov/media/91668/download Available at:

28. Stolley P.D. Asthma mortality: why the United States was spared an epidemic of deaths due to asthma. Am Rev Respir Dis. 1972;105:883–890. [PubMed] [Google Scholar]

29. Schardein J. Vol. 5. CRC Printing; Cleveland, OH: 1976. Drugs as Teratogens; p. 49. [Google Scholar]

xxx. Suntharalingam G., Perry M.R., Ward Due south. Cytokine storm in stage i trial of the anti-CD28 monoclonal antibody TGN1312. Due north Engl J Med. 2006;355:1018–1028. [PubMed] [Google Scholar]

31. Dayan C.M., Wraith D.C. Preparing for first-in-man studies: the challenges for translational immunology postal service TGN1412. Clin Exp Immunol. 2008;151:231–234. [PMC free commodity] [PubMed] [Google Scholar]

34. Vial T., Descotes J., Braun F., Behrend Yard. Affiliate 37. Drugs that act on the immune system: 37—--cytokines and monoclonal antibodies. Side Effects Drugs Annu. 2007;29:383–423. [Google Scholar]

35. Eddleston M., Cohen A.F., Webb D.J. Implications of the BIA-102474-101 written report for review of first-into-human clinical trials. Brit J Clin Pharm. 2016;81:582–586. [PMC free article] [PubMed] [Google Scholar]

36. Sankar U. The frail toxicity balance in drug discovery. Scientist. 2005;19:32. [Google Scholar]

37. Gura T. Systems for identifying new drugs are oft faulty. Science. 1997;273:1041–1042. [PubMed] [Google Scholar]

38. Koppanyi T., Avery M.A. Species differences and the clinical trial of new drugs: a review. Clin Pharmacol Ther. 1966;7:250–270. [PubMed] [Google Scholar]

39. Cormia F.E., Lewis G.M., Hopper Grand.E. Toxicity of penicillin for the guinea squealer. J Invest Dermatol. 1947;9:261–267. [PubMed] [Google Scholar]

xl. Villar D., Buck W.B., Gonzalez J.G. Ibuprofen, aspirin and acetaminophen toxicosis and treatment in dogs and cats. Vet Human Toxicol. 1998;40:156–162. [PubMed] [Google Scholar]

41. Wilson J.G., Ritter E.J., Scott W.J., Fradkin R. Comparative distribution and embryotoxicity of acetylsalicylic acid in pregnant rats and rhesus monkeys. Toxicol Appl Pharmacol. 1977;41:67–78. [PubMed] [Google Scholar]

42. Buhles W.C. Compassionate use: a story of ideals and science in the development of a new drug. In: Monticello M., editor. Vol. 54. Johns Hopkins University Press; Baltimore, MD: 2011. pp. 304–315. (Perspectives and Biology in Medicine). [PubMed] [Google Scholar]

45. Meigs L., Smirova L., Rovida C., Leist M., Hartung T. Animal testing and its alternatives—the most important omics is economic science. ALTEX. 2018;35:275–305. [PubMed] [Google Scholar]

46. USDA National Agronomical Library, U.South Department of Agriculture. Animal Welfare Act. USDA National Agricultural Library, U.S. Section of Agriculture. https://www.nal.usda.gov/awic/animal-welfare-act Available at:

47. Taylor K., Gordon N., Langley G., Higgins W. Estimates for worldwide laboratory beast utilize in 2005. Altern Lab Anim. 2008;36:327–342. [PubMed] [Google Scholar]

49. Keen J. Wasted coin in United States biomedical and agricultural beast research. In: Hermann Yard., Jayne Thou., editors. Vol 22. Brill; Boston, MA: 2019. pp. 244–272. (Creature Experimentation: Working Towards a Paradigm Change). [Google Scholar]

50. Browne P., Judson R.S., Casey W.Chiliad., Kleinstreuer N.C., Thomas R.S. Screening chemicals for estrogen receptor bioactivity using a computations model. Environ Sci Technol. 2015;49:8804–8814. [PubMed] [Google Scholar]

52. Bailey J., Thew Yard., Balls M. An assay of the use of dogs in predicting human toxicology and drug safety. Altern Lab Anim. 2013:335–350. [PubMed] [Google Scholar]

54. Grimes D.A., Schultz G.F. Refining clinical diagnosis with likelihood ratios. Lancet. 2005;365:1500–1505. [PubMed] [Google Scholar]

55. Matthews R.A. Medical progress depends on beast models—doesn't information technology? J R Soc Med. 2008;101:95–98. [PMC complimentary article] [PubMed] [Google Scholar]

56. Paglialunga Southward., Morimoto B.H., Clark Yard., Friedrichs G.Southward. Translatability of the S7A cadre battery respiratory safety pharmacology studies: preclinical respiratory and related clinical adverse events. J Pharmacol Toxicol Methods. 2019 Jun v [E-pub ahead of print] [PubMed] [Google Scholar]

57. Lichtfield J.T., Jr. Symposium on clinical drug evaluation and human pharmacology. Sixteen. Evaluation of the condom of new drugs past means of tests in animals. Clin Pharmacol Ther. 1962;3:665–672. [PubMed] [Google Scholar]

58. Lumley C. Clinical toxicity: could it have been predicted? Premarketing feel. In: Lumley C., Walker Due south., editors. Animal Toxicity Studies: Their Relevance for Man. Quay Publishing; London, Britain: 1990. pp. 49–56. [Google Scholar]

59. Council on Scientific Affairs Carcinogen regulation. JAMA. 1981;246:253–256. [PubMed] [Google Scholar]

sixty. Abi-Gerges N., McMahon C., Vargas H. The west coast regional safety pharmacology club meeting update: filling in the translational gaps in safety assessment. J Pharmacol Toxicol Methods. 2019;98:106582. [PubMed] [Google Scholar]

61. Ewert L., Aylott 1000., Deurinck One thousand. The concordance between nonclinical and phase I clinical cardiovascular assessment from a cross-company data sharing initiative. Toxicol Sci. 2014;142:427–435. [PubMed] [Google Scholar]

62. Shanks N., Greek R., Greek J. Are creature models predictive for humans? Philos Ethics Humanit Med. 2009;4:two. [PMC costless article] [PubMed] [Google Scholar]

63. U.S. Nutrient and Drug Administration . U.S. Food and Drug Administration; Washington DC: 2006. Press release: FDA issues advice to make earliest stages of clinical drug evolution more efficient. January 12. [Google Scholar]

64. Plant of Medicine Committee on the Use of Chimpanzees in Biomedical and Behavioral Research . National Academic Press; Washington, DC: 2011. Chimpanzees in Biomedical and Behavioral Inquiry: Assessing the Necessity. [Google Scholar]

67. Bracken M.B. Why beast studies are often poor predictors of human reactions to exposure. J R Soc Med. 2009;102:120–122. [PMC complimentary article] [PubMed] [Google Scholar]

68. van der Worp H.B., Howells D.West., Sena Eastward.Southward. Can creature models of disease reliably inform human studies? PLoS Med. 2010;7 [PMC gratis article] [PubMed] [Google Scholar]

69. Greek R., Kramer L.A. How to evaluate the scientific discipline of non-homo fauna use in biomedical research and testing: a proposed format for argue. In: Herrmann K., Jayne Thou., editors. Vol. 22. Brill Open; Boston, MA: 2019. pp. 65–87. (Animate being Experimentation: Working Towards a Paradigm of Alter). [Google Scholar]

seventy. Ioannidis J.P.A. Extrapolating from animals to humans. Sci Transl Med. 2012;4:151. [PubMed] [Google Scholar]

71. Greek R., Menache A. Systematic reviews of beast models: methodology versus epistemology. Int J Med Sci. 2013;10:206–221. [PMC free article] [PubMed] [Google Scholar]

72. Knight A. Systematic reviews of fauna experiments demonstrate poor human clinical and toxicological utility. Altern Lab Anim. 2007;35:641–659. [PubMed] [Google Scholar]

73. Akhtar A. The flaws and human being harms of brute experimentation. Camb Q Healthc Ethics. 2015;24:407–419. [PMC free article] [PubMed] [Google Scholar]

74. Pound P., Ritskes-Hoiting M. Is it possible to overcome issues of external validity in preclinical animal research? Why well-nigh brute models are bound to fail. J Transl Med. 2018;16:304. [PMC free article] [PubMed] [Google Scholar]

75. Miquel P.A. Extended physics as a theoretical framework for systems biology? Prog Biophys Mol Biol. 2011;106:348–352. [PubMed] [Google Scholar]

76. Soto A.M., Sonnenschein C., Maini P.K., Novle D. Systems biology and cancer. Prog Biophys Mol Biol. 2011;106:337–346. [PMC gratuitous commodity] [PubMed] [Google Scholar]

77. Groell F., Jordan O., Borchard 1000. In vitro models for immunogenicity prediction of therapeutic proteins. Eur J Pharm Biopharm. 2018;130:128–142. [PubMed] [Google Scholar]

78. Mittal R., Woo F.W., Castro C.Southward. Organ-on-chip models: implications in drug discovery and clinical applications. J Cell Physiol. 2019;234:8352–8380. [PubMed] [Google Scholar]

79. Benam K.H., Gilchrist S., Kleensang A., Satz A.B., Willet C., Zhang Q. Exploring new technologies in biomedical research. Drug Discov Today. 2019;24:1242–1247. [PubMed] [Google Scholar]

lxxx. Doke S.K., Dhawale S.C. Alternatives to animal testing: a review. Saudi Pharm J. 2015;23:223–229. [PMC free article] [PubMed] [Google Scholar]

81. Burt T., Vuong Fifty.T., Baker E. Phase 0, including microdosing approaches: applying the iii Rs and increasing the efficiency of homo drug development. Altern Lab Anim. 2018;46 335–four. [PubMed] [Google Scholar]

82. Herrmann K., Pistollato F., Stephens M.Fifty. Across the iii Rs: expanding the use of human-relevant replacement methods in biomedical enquiry. ALTEX. 2019;36:343–352. [PubMed] [Google Scholar]


Articles from JACC: Basic to Translational Scientific discipline are provided hither courtesy of Elsevier


Source: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6978558/

Posted by: ortizfirembles.blogspot.com

0 Response to "Why Do We Use Animals In Toxicological Studies, Instead Of Humans?"

Post a Comment

Iklan Atas Artikel

Iklan Tengah Artikel 1

Iklan Tengah Artikel 2

Iklan Bawah Artikel