Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 548
Filter
1.
AMA J Ethics ; 26(9): E679-683, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39250938

ABSTRACT

Nonhuman animals used in biomedical research frequently suffer and are harmed as part of their use as experimental models. The Institutional Animal Care and Use Committee (IACUC) of a given institution is meant to ensure that research protocols follow federal guidelines, but research protocols such as those described in this case can generate unnecessary suffering; this problem suggests limitations of IACUCs' capacity to protect nonhuman animals' welfare. This commentary on the case considers how to more fully protect nonhuman animals used in scientific research and identifies barriers to more comprehensive protection of nonhuman animals' welfare.


Subject(s)
Animal Care Committees , Animal Experimentation , Animal Welfare , Animals, Laboratory , Animal Welfare/ethics , Animal Welfare/standards , Animals , Animal Experimentation/ethics , Animal Experimentation/standards , Biomedical Research/ethics , Biomedical Research/standards , Guidelines as Topic , Humans , United States , Ethics, Research
2.
Regul Toxicol Pharmacol ; 152: 105683, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39117168

ABSTRACT

Following the European Commission decision to develop a roadmap to phase out animal testing and the signing of the US Modernisation Act, there is additional pressure on regulators and the pharmaceutical industry to abandon animal experimentation in safety testing. Often, endeavours already made by governments, regulators, trade associations, and industry to replace, reduce and refine animal experimentation (3Rs) are unnoticed. Herein, we review such endeavours to promote wider application and acceptance of 3Rs. ICH guidelines have stated 3Rs objectives and have enjoyed many successes driven by global consensus. Initiatives driven by US and European regulators such as the removal of the Abnormal Toxicity Test are neutralised by reticent regional regulators. Stream-lined testing requirements have been proposed for new modalities, oncology, impurity management and animal pharmacokinetics/metabolism. Use of virtual controls, value of the second toxicity species, information sharing and expectations for life-threatening diseases, human specific or well-characterised targets are currently being scrutinised. Despite much effort, progress falls short of the ambitious intent of decisionmakers. From a clinical safety and litigation perspective pharmaceutical companies and regulators are reluctant to step away from current paradigms unless replacement approaches are validated and globally accepted. Such consensus has historically been best achieved through ICH initiatives.


Subject(s)
Animal Testing Alternatives , Drug Industry , Toxicity Tests , Animals , Drug Industry/standards , Drug Industry/legislation & jurisprudence , Humans , Animal Experimentation/standards , Pharmaceutical Preparations/standards , Drug-Related Side Effects and Adverse Reactions
4.
Nat Commun ; 15(1): 5574, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956430

ABSTRACT

The biomedical research community addresses reproducibility challenges in animal studies through standardized nomenclature, improved experimental design, transparent reporting, data sharing, and centralized repositories. The ARRIVE guidelines outline documentation standards for laboratory animals in experiments, but genetic information is often incomplete. To remedy this, we propose the Laboratory Animal Genetic Reporting (LAG-R) framework. LAG-R aims to document animals' genetic makeup in scientific publications, providing essential details for replication and appropriate model use. While verifying complete genetic compositions may be impractical, better reporting and validation efforts enhance reliability of research. LAG-R standardization will bolster reproducibility, peer review, and overall scientific rigor.


Subject(s)
Animals, Laboratory , Guidelines as Topic , Animals , Animals, Laboratory/genetics , Reproducibility of Results , Research Design , Animal Experimentation/standards , Biomedical Research/standards
5.
Eur J Orthod ; 46(4)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39007674

ABSTRACT

BACKGROUND: The ARRIVE 2.0 guidelines were introduced to improve the reporting of animal studies. The aim of this study was to assess the reporting adherence of orthodontic speciality animal studies in relation to ARRIVE 2.0 guidelines. Associations between the reporting and study characteristics were explored. MATERIALS AND METHOD: An electronic database search was undertaken using Medline via PubMed (www.pubmed.ncbi.nlm.nih.gov) to identify studies meeting the eligibility criteria published between 1 January 2018 and 31 December 2023. Data extraction was performed in duplicate and independently. Descriptive statistics and frequency distributions for the responses to each checklist item were calculated. Mean values for adequate reporting per ARRIVE item were calculated. A sum score was calculated by adding the responses (0 = not reported, 1 = inadequate reporting, 2 = adequate reporting) per item and sub-questions. On an exploratory basis, univariable linear regression between summary score and study characteristics (year of publication, continent of authorship, type of centre, and number of authors) was performed. RESULTS: Three hundred and eighty-four studies were analysed. Variability in the adequate reporting of the ARRIVE 2.0 guideline items was evident. In particular, in 32% of studies, there was a lack of reporting of the priori sample size calculation. Overall, the mean reporting score for the sample was 57.9 (SD 6.7 and range 34-74). There were no associations between score and study characteristics except for a weak association for year of publication with a small improvement over time (each additional year). CONCLUSIONS: The reporting of animal studies relevant to the speciality of orthodontics is sub-optimal in relation to the ARRIVE 2.0 guidelines. There was a tendency for the non-reporting of items pertaining to study sample size, eligibility, methods to reduce bias and interpretation/scientific implications. Greater awareness and reporting adherence to the ARRIVE 2.0 guidelines are required to reduce research waste involving animal models.


Subject(s)
Orthodontics , Orthodontics/standards , Animals , Models, Animal , Research Design/standards , Guidelines as Topic , Checklist , Guideline Adherence , Animal Experimentation/standards , Dental Research/standards , Publishing/standards
7.
J Am Assoc Lab Anim Sci ; 63(3): 209-220, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38749659

ABSTRACT

Animal research facilities are noisy environments. The high air change rates required in animal housing spaces tend to create higher noise levels from the heating and cooling systems. Housing rooms are typically constructed of hard wall material that is easily cleaned but simultaneously highly reverberant, meaning that the sound cannot be controlled/attenuated so the sounds that are generated bounce around the room uncontrolled. (Soft, sound-absorbing surfaces generally cannot be used in animal facilities because they collect microbes; various wall surface features and sound control panel options are available, although rarely used.) In addition, many of our husbandry tasks such as cage changing, animal health checks, cleaning, and transporting animals produce high levels of noise. Finally, much of the equipment we have increasingly employed in animal housing spaces, such as ventilated caging motors, biosafety, or procedure cabinets, can generate high levels of background noise, including ultrasound. These and many additional factors conspire to create an acoustic environment that is neither naturalistic nor conducive to a stress-free environment. The acoustic variability both within and between institutions can serve as an enormous confounder for research studies and a threat to our ability to reproduce studies over time and between research laboratories. By gaining a better appreciation for the acoustic variables, paired with transparency in reporting the levels, we might be able to gain a better understanding of their impacts and thereby gain some level of control over such acoustic variables in the animal housing space. The result of this could improve both animal welfare and study reproducibility, helping to address our 3Rs goals of Replacement, Reduction, and Refinement in the animal biomedical research enterprise.


Subject(s)
Animal Husbandry , Animals, Laboratory , Housing, Animal , Noise , Noise/adverse effects , Animals , Animal Husbandry/methods , Animal Welfare , Animal Experimentation/standards
8.
ALTEX ; 41(3): 382-394, 2024.
Article in English | MEDLINE | ID: mdl-38492209

ABSTRACT

Following a review of Directive 2010/63/EU on the protection of animals used for scientific pur­poses in the European Union (EU), non-technical project summaries (NTS) of all approved projects must be published in a central database using a standard template. Our initial review of the NTS reported in ALTEX in 2018 had found the NTS to be deficient in their accessibility and quality, notably the "adverse effects" section where the harms to the animals are meant to be described. Here we repeat our review to see if these legislative changes have improved the accessibility and quality of the NTS. As before, we focused on the NTS from the United Kingdom (UK) and Germany; even though the UK has left the EU, it is using the same template. We found significant improvement in the reporting of five of the six elements we identified as essential to the "predicted harms" section. However, there was no significant improvement in the reporting of adverse effects. Only 41% of German NTS and 48% of UK NTS are fully reporting this important element of the "predicted harms" section. In our view, researchers need support in describing the impact of their research on the animals and to assist here we include a checklist for competent authorities and a list of suggested terminology for standard administration and sampling procedures. Unless the NTS improve further, their utility as a tool for sharing of good practices in the 3Rs or to support evidence-based policy­making will remain limited.


All countries of the European Union (EU) are required to publish "non-technical summaries" (NTS) of research projects that use animals. To improve transparency, the public must have access to NTS and understand their content. Our previous review found that the information provided in the NTS was lacking in many cases. This is preventing a full understanding of what animals experience during experiments. In particular, NTS often failed to fully describe what procedures the animals would be subjected to, how often they would take place, how long they would last, and the harm they would cause. Here we repeat our review to see if recent legislative changes, including the requirement for NTS to be published in a central database using a standard template, have made a difference. While there has been some improvement in reporting, many NTS still fail to adequately describe the harm that animals will experience.


Subject(s)
Animal Experimentation , Animal Testing Alternatives , Animals , Animal Testing Alternatives/legislation & jurisprudence , Animal Experimentation/legislation & jurisprudence , Animal Experimentation/standards , Europe , Animal Welfare/legislation & jurisprudence , Animal Welfare/standards , European Union
9.
J Am Assoc Lab Anim Sci ; 63(2): 107-115, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38182133

ABSTRACT

Vibration is inherent in research animal facilities due to the mechanical systems and practices required for animal care and use. Ample evidence indicates that vibration can change behavior and physiology in multiple species, potentially altering the results of research studies. Although one cannot eliminate environmental vibration, its control is important in research animal environments to decrease the possibility of introducing a research variable due to vibration effects. To assess the potential for a vibration source to alter experimental results and variability, one must understand the principles of vibration, its likely sources, and control methods. The literature regarding the effects of vibration, as it applies in a practical sense, can be challenging to interpret because the vibration frequencies tested to date have often not been within or near the most sensitive ranges of the species being tested. Some previous studies have used unrealistic vibration magnitudes and provided insufficient detail to duplicate or build upon conclusions. Standardization is essential for research examining the effects of vibration on animals to validate knowledge of this extrinsic variable in animal research and identify ways to mitigate the variable in research facilities.


Subject(s)
Animals, Laboratory , Vibration , Vibration/adverse effects , Animals , Animals, Laboratory/physiology , Animal Experimentation/standards
10.
PLoS One ; 17(11): e0275962, 2022.
Article in English | MEDLINE | ID: mdl-36327216

ABSTRACT

Lack of translation and irreproducibility challenge preclinical animal research. Insufficient reporting methodologies to safeguard study quality is part of the reason. This nationwide study investigates the reporting prevalence of these methodologies and scrutinizes the reported information's level of detail. Publications were from two time periods to convey any reporting progress and had at least one author affiliated to a Danish University. We retrieved all relevant animal experimental studies using a predefined research protocol and a systematic search. A random sampling of 250 studies from 2009 and 2018 led to 500 publications in total. Reporting of measures known to impact study results estimates were assessed. Part I discloses a simplified two-level scoring "yes/no" to identify the presence of reporting. Part II demonstrates an additional three-level scoring to analyze the reported information's level of detail. Overall reporting prevalence is low, although minor improvements are noted. Reporting of randomization increased from 24.0% in 2009 to 40.8% in 2018, blinded experiment conduct from 2.4% to 4.4%, blinded outcome assessment from 23.6% to 38.0%, and sample size calculation from 3.2% to 14.0%. Poor reporting of details is striking with reporting of the random allocation method to groups being only 1.2% in 2009 and 6.0% in 2018. Reporting of sample size calculation method was 2.4% in 2009 and 7.6% in 2018. Only conflict-of-interest statements reporting increased from 37.6% in 2009 to 90.4%. Measures safeguarding study quality are poorly reported in publications affiliated with Danish research institutions. Only a modest improvement was noted during the period 2009-2018, and the lack of details urgently prompts institutional strategies to accelerate this. We suggest thorough teaching in designing, conducting and reporting animal studies. Education in systematic review methodology should be implemented in this training and will increase motivation and behavior working towards quality improvements in science.


Subject(s)
Animal Experimentation , Research Design , Animals , Animal Experimentation/standards , Quality Improvement , Research Design/standards
11.
PLoS One ; 16(12): e0260114, 2021.
Article in English | MEDLINE | ID: mdl-34851985

ABSTRACT

One response to calls for increased openness in animal research is to make protocols publicly accessible, but it is unclear what type of input the public would provide if given this opportunity. In this study we invited public responses to five different research projects, using non-technical summaries intended for lay audiences. Our aim was to assess the potential for this type of public consultation in protocol review, and a secondary aim was to better understand what types of animal research people are willing to accept and why. US participants (n = 1521) were asked (via an online survey) "Do you support the use of these (insert species) for this research", and responded using a seven-point scale (1 = "No", 4 = "Neutral", and 7 = "Yes"). Participants were asked to explain the reasons for their choice; open-ended text responses were subjected to thematic analysis. Most participants (89.7%) provided clear comments, showing the potential of an online forum to elicit feedback. Four themes were prevalent in participant reasoning regarding their support for the proposed research: 1) impact on animals, 2) impact on humans, 3) scientific merit, and 4) availability of alternatives. Participant support for the proposed research varied but on average was close to neutral (mean ± SD: 4.5 ± 2.19) suggesting some ambivalence to this animal use. The protocol describing Parkinson's research (on monkeys) was least supported (3.9 ± 2.17) and the transplant research (on pigs) was most supported (4.9 ± 2.02). These results indicate that public participants are sensitive to specifics of a protocol. We conclude that an online forum can provide meaningful public input on proposed animal research, offering research institutions the opportunity for improved transparency and the chance to reduce the risk that they engage in studies that are out of step with community values.


Subject(s)
Animal Experimentation/ethics , Animal Welfare/ethics , Public Opinion , Animal Experimentation/standards , Animal Welfare/standards , Animals , Attitude , Guideline Adherence , Humans , Practice Guidelines as Topic
12.
PLoS Biol ; 19(9): e3001397, 2021 09.
Article in English | MEDLINE | ID: mdl-34499640

ABSTRACT

In 2018, the first registry dedicated to preregistration of animal study protocols was launched. Despite international support, the overall number of (pre)registered protocols is still low, illustrating the need for pushing the preregistration agenda among researchers and policymakers.


Subject(s)
Registries , Research Design , Animal Experimentation/standards , Animals
13.
Gut Microbes ; 13(1): 1941711, 2021.
Article in English | MEDLINE | ID: mdl-34328058

ABSTRACT

In recent years, studies investigating the role of the gut microbiota in health and diseases have increased enormously - making it essential to deepen and question the research methodology employed. Fecal microbiota transplantation (FMT) in rodent studies (either from human or animal donors) allows us to better understand the causal role of the intestinal microbiota across multiple fields. However, this technique lacks standardization and requires careful experimental design in order to obtain optimal results. By comparing several studies in which rodents are the final recipients of FMT, we summarize the common practices employed. In this review, we document the limitations of this method and highlight different parameters to be considered while designing FMT Studies. Standardizing this method is challenging, as it differs according to the research topic, but avoiding common pitfalls is feasible. Several methodological questions remain unanswered to this day and we offer a discussion on issues to be explored in future studies.


Subject(s)
Animal Experimentation/standards , Fecal Microbiota Transplantation/standards , Feces/microbiology , Gastrointestinal Microbiome , Guidelines as Topic , Rodentia/microbiology , Animals , Disease Models, Animal , Germ-Free Life , Humans
14.
Exp Anim ; 70(4): 532-540, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34193732

ABSTRACT

The Institutional Animal Care and Use Committee (IACUC) of Seoul National University (SNU) plays a key role in monitoring and managing the humane use of animals in scientific research. Here, as one of the pioneers of the IACUC in Korea, we reported SNU-IACUC operations and activities including committee establishment and legal formulation, protocol review, and post-approval monitoring of protocols, which the IACUC has undertaken in the last decade. In addition, legal regulations and improvements were also discussed, and encompassed the limited number of committee members and the single IACUC policy in Korea. As of December, 2020, amendments are on the table at the National Assembly. We also emphasized the independent nature of the IACUC in protecting activities, including approval and monitoring animal experiments, and its public role in narrowing the knowledge gap between society and scientists. Thus, the aim of this report is to help society and scientists understand the operations of the SNU-IACUC and its role in animal welfare.


Subject(s)
Animal Care Committees/history , Animal Experimentation/standards , Animal Welfare/standards , Animals, Laboratory , Animals , History, 21st Century , Seoul , Universities
15.
Lab Anim ; 55(3): 244-253, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34085570

ABSTRACT

Appropriate end-points are integral to the refinement of laboratory animal experiments. Our recent experience has highlighted that ambiguity around end-points is hampering their adoption in experiments that cause severe suffering to fish. In toxicology, the term endpoint (single word) refers to the response variable to the treatment that is measured and analysed. This differs to usage within laboratory animal experimentation, where end-point (hyphenated) refers to the time-point when exposure of the animal(s) to the treatment (and suffering) ends. Within laboratory animal experimentation, standardised terminology is needed for different types of early end-point which are based on the condition of the animal(s) or progress of the experiment. We propose that those involved in regulating and conducting animal experiments consider seven distinct types of early end-point (aim, technical error, biological error, mortality, moribundity, prognostic humane, non-prognostic humane) in addition to the planned experimental end-point (i.e. maximum duration). Moribundity (not morbidity) refers to an animal in a severely debilitated state close to death. Moribundity in fish is not yet defined, so we propose identification via a lack of response to external stimuli, loss of equilibrium (i.e. loss of righting reflex), and a slow opercular ventilation rate. As these clinical signs equate to those of deep/surgical anaesthesia, this moribundity end-point cannot be considered a humane end-point as the fish is likely to be unconscious and have passed the point of maximum suffering. We believe that identification of earlier humane end-points based on clinical signs and wider recognition of other types of early end-point can reduce suffering in experiments.


Subject(s)
Animal Experimentation/standards , Animals, Laboratory , Fishes , Research Design/standards , Animals
16.
Regul Toxicol Pharmacol ; 123: 104953, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33984412

ABSTRACT

The safety testing of pharmaceutical candidates has traditionally relied on data gathered from studies in animals, and these sources of information remain a vital component of the safety assessment for new drug and biologic products. However, there are clearly ethical implications that attend the use of animals for safety testing, and FDA fully supports the principles of the 3Rs, as it relates to animal usage; these being to replace, reduce and refine. We provide an overview of some of the events and activities (legal and programmatic) that have had, and continue to have, the greatest impact on animal use in pharmaceutical development, and highlight some ongoing efforts to further meet the challenge of achieving our mission as humanely as possible.


Subject(s)
Animal Experimentation , Animal Experimentation/standards , Animal Testing Alternatives , Animal Welfare , Animals
17.
PLoS Biol ; 19(5): e3001009, 2021 05.
Article in English | MEDLINE | ID: mdl-34010281

ABSTRACT

The replicability of research results has been a cause of increasing concern to the scientific community. The long-held belief that experimental standardization begets replicability has also been recently challenged, with the observation that the reduction of variability within studies can lead to idiosyncratic, lab-specific results that cannot be replicated. An alternative approach is to, instead, deliberately introduce heterogeneity, known as "heterogenization" of experimental design. Here, we explore a novel perspective in the heterogenization program in a meta-analysis of variability in observed phenotypic outcomes in both control and experimental animal models of ischemic stroke. First, by quantifying interindividual variability across control groups, we illustrate that the amount of heterogeneity in disease state (infarct volume) differs according to methodological approach, for example, in disease induction methods and disease models. We argue that such methods may improve replicability by creating diverse and representative distribution of baseline disease state in the reference group, against which treatment efficacy is assessed. Second, we illustrate how meta-analysis can be used to simultaneously assess efficacy and stability (i.e., mean effect and among-individual variability). We identify treatments that have efficacy and are generalizable to the population level (i.e., low interindividual variability), as well as those where there is high interindividual variability in response; for these, latter treatments translation to a clinical setting may require nuance. We argue that by embracing rather than seeking to minimize variability in phenotypic outcomes, we can motivate the shift toward heterogenization and improve both the replicability and generalizability of preclinical research.


Subject(s)
Animal Experimentation/standards , Research Design/standards , Animals , Behavior, Animal/physiology , Brain Ischemia/metabolism , Humans , Meta-Analysis as Topic , Models, Animal , Phenotype , Reference Standards , Reproducibility of Results , Research Design/trends , Stroke/physiopathology
18.
Front Endocrinol (Lausanne) ; 12: 638261, 2021.
Article in English | MEDLINE | ID: mdl-34040580

ABSTRACT

A well-documented method and experimental design are essential to ensure the reproducibility and reliability in animal research. Experimental studies using exercise programs in animal models have experienced an exponential increase in the last decades. Complete reporting of forced wheel and treadmill exercise protocols would help to ensure the reproducibility of training programs. However, forced exercise programs are characterized by a poorly detailed methodology. Also, current guidelines do not cover the minimum data that must be included in published works to reproduce training programs. For this reason, we have carried out a systematic review to determine the reproducibility of training programs and experimental designs of published research in rodents using a forced wheel system. Having determined that most of the studies were not detailed enough to be reproducible, we have suggested guidelines for animal research using FORCED exercise wheels, which could also be applicable to any form of forced exercise.


Subject(s)
Animal Experimentation/standards , Disease Models, Animal , Exercise Test , Physical Conditioning, Animal , Animals , Exercise , Female , Humans , Humidity , Male , Mice , Rats , Reproducibility of Results , Risk , Temperature
19.
PLoS Biol ; 19(5): e3001177, 2021 05.
Article in English | MEDLINE | ID: mdl-33951050

ABSTRACT

In an effort to better utilize published evidence obtained from animal experiments, systematic reviews of preclinical studies are increasingly more common-along with the methods and tools to appraise them (e.g., SYstematic Review Center for Laboratory animal Experimentation [SYRCLE's] risk of bias tool). We performed a cross-sectional study of a sample of recent preclinical systematic reviews (2015-2018) and examined a range of epidemiological characteristics and used a 46-item checklist to assess reporting details. We identified 442 reviews published across 43 countries in 23 different disease domains that used 26 animal species. Reporting of key details to ensure transparency and reproducibility was inconsistent across reviews and within article sections. Items were most completely reported in the title, introduction, and results sections of the reviews, while least reported in the methods and discussion sections. Less than half of reviews reported that a risk of bias assessment for internal and external validity was undertaken, and none reported methods for evaluating construct validity. Our results demonstrate that a considerable number of preclinical systematic reviews investigating diverse topics have been conducted; however, their quality of reporting is inconsistent. Our study provides the justification and evidence to inform the development of guidelines for conducting and reporting preclinical systematic reviews.


Subject(s)
Peer Review, Research/methods , Peer Review, Research/standards , Research Design/standards , Animal Experimentation/standards , Animals , Bias , Checklist/standards , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Empirical Research , Epidemiologic Methods , Epidemiology/trends , Humans , Peer Review, Research/trends , Publications , Reproducibility of Results , Research Design/trends
20.
Lab Anim ; 55(3): 233-243, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33215575

ABSTRACT

Using animals for research raises ethical concerns that are addressed in project evaluation by weighing expected harm to animals against expected benefit to society. A harm-benefit analysis (HBA) relies on two preconditions: (a) the study protocol is scientifically suitable and (b) the use of (sentient) animals and harm imposed on them are necessary for achieving the study's aims. The 3Rs (Replace, Reduce and Refine) provide a guiding principle for evaluating whether the use of animals, their number and the harm imposed on them are necessary. A similar guiding principle for evaluating whether a study protocol is scientifically suitable has recently been proposed: the 3Vs principle referring to the three main aspects of scientific validity in animal research (construct, internal and external validity). Here, we analyse the internal consistency and compatibility of these two principles, address conflicts within and between the 3Rs and 3Vs principles and discuss their implications for project evaluation. We show that a few conflicts and trade-offs exist, but that these can be resolved either by appropriate study designs or by ethical deliberation in the HBA. In combination, the 3Vs, 3Rs and the HBA thus offer a coherent framework for a logically structured evaluation procedure to decide about the legitimacy of animal research projects.


Subject(s)
Animal Experimentation/standards , Animal Testing Alternatives/statistics & numerical data , Research Design/standards
SELECTION OF CITATIONS
SEARCH DETAIL