Animal Testing and Drug Development Research Paper

Academic Writing Service

Sample Animal Testing and Drug Development Research Paper. Browse other research paper examples and check the list of argumentative research paper topics for more inspiration. If you need a research paper written according to all the academic standards, you can always turn to our experienced writers for help. This is how your paper can get an A! Also, chech our custom research proposal writing service for professional assistance. We offer high-quality assignments for reasonable rates.

This research paper examines the intricate relationship between animal testing and drug development, focusing on the efficacy and safety aspects of this longstanding practice. It navigates through the historical evolution of animal testing, the ethical debates surrounding its use, and the pivotal role it plays in predicting drug efficacy and safety for human applications. By reviewing case studies and highlighting both its successes and limitations, this study underscores the essential contributions of animal testing to pharmaceutical advancements while acknowledging the ethical dilemmas it raises. It also explores emerging alternatives and ethical considerations, shedding light on the evolving landscape of drug testing methodologies. Ultimately, this research elucidates the complex interplay of science, ethics, and innovation within the realm of animal testing and drug development, offering valuable insights for future research and policy considerations in this field.

Academic Writing, Editing, Proofreading, And Problem Solving Services

Get 10% OFF with 24START discount code


I. Introduction

Background Information

The historical employment of animals in drug development traces its roots to ancient civilizations, where empirical observations on animals’ responses to various substances laid the foundation for pharmacological studies. However, it was in the 19th century that systematic animal testing gained prominence with the advent of controlled experiments and the isolation of active compounds. The subsequent decades witnessed the refinement of animal models and the establishment of regulatory frameworks mandating their use in safety and efficacy assessments for pharmaceuticals. These historical developments underscore the enduring significance of animal testing in the pharmaceutical industry, despite evolving scientific methodologies and ethical debates surrounding its practice (Kirk, 2018; Franco et al., 2020).

Problem Statement

The ethical concerns and controversies surrounding animal testing have become increasingly prominent in contemporary discourse. Critics argue that it raises profound moral and welfare issues, as animals subjected to experimentation endure suffering and often face adverse consequences. The ethical dilemma revolves around balancing potential benefits to human health with the welfare of sentient creatures. Furthermore, there are questions about the predictive validity of animal models in determining drug safety and efficacy for humans, which has prompted calls for alternative testing methods that are both ethically sound and scientifically robust (Ormandy et al., 2019; Pound and Ritskes-Hoitinga, 2018).




Purpose of the Paper

This research paper aims to comprehensively investigate the interplay between animal testing and drug development, with a particular focus on evaluating its efficacy and safety aspects. By critically examining historical and contemporary practices, ethical considerations, and emerging alternatives, the primary objective is to provide a nuanced understanding of the complex dynamics that define this critical facet of biomedical research.

Research Questions

The specific questions guiding this research include:

  1. To what extent has animal testing historically contributed to drug development and safety assessments?
  2. What are the key ethical concerns and controversies associated with animal testing in the pharmaceutical industry?
  3. How do animal models compare to alternative testing methods in predicting drug efficacy and safety for humans?
  4. What are the implications of evolving ethical and scientific considerations for the future of animal testing in drug development?

Significance of the Study

Understanding the efficacy and safety of animal testing in drug development holds paramount importance in modern society. The pharmaceutical industry heavily relies on animal models to ensure drug safety and effectiveness before human trials, making this research pertinent to public health. Additionally, this study addresses the pressing ethical concerns surrounding animal welfare and seeks to contribute to the ongoing discourse on responsible and sustainable research practices. By shedding light on the complexities of this issue, it informs policymakers, researchers, and the general public about the multifaceted nature of animal testing in the context of drug development, thus fostering informed decision-making and ethical progress in the field (Akhtar, 2015; Huxtable, 2020).

II. Literature Review

Historical Evolution of Animal Testing

The historical evolution of animal testing in drug development reflects a trajectory marked by both scientific advancements and evolving ethical norms. From its rudimentary origins in ancient Greece, where observations on animal reactions to substances laid the groundwork for pharmacological inquiries, to the structured experiments of the 19th century, animal testing gradually became a standardized practice. The establishment of regulatory frameworks in the 20th century, such as the U.S. Food and Drug Administration’s (FDA) requirements for animal testing in drug safety assessments, institutionalized its role in pharmaceutical research (Kirk, 2018). Over time, this evolution has seen the refinement of animal models and an expansion of the types of animals used, as well as heightened scrutiny of its ethical implications.

Ethical Considerations

Ethical debates surrounding animal testing have intensified in recent years, fueled by growing concerns about animal welfare and the moral consequences of experimentation. Advocates argue that animal testing is essential for advancing medical knowledge and drug development, potentially leading to life-saving therapies. Conversely, critics contend that it raises profound ethical dilemmas, as animals experience suffering and harm during experiments. The ethical considerations involve a delicate balance between the potential benefits to human health and the well-being of sentient creatures, with some advocating for stricter regulations, improved laboratory conditions, and a shift toward alternative methods that do not involve animals (Ormandy et al., 2019; Pound and Ritskes-Hoitinga, 2018).

Animal Models in Drug Development

Commonly used animals in drug testing include rodents (mice and rats), rabbits, dogs, and non-human primates, each chosen based on their suitability for specific research objectives. Rodents, owing to their genetic similarity to humans and ease of handling, are frequently employed for preliminary testing, while dogs and non-human primates may be used for assessing drug safety and pharmacokinetics. However, despite their widespread use, these models have inherent limitations. Variations in physiology, metabolism, and genetics between animals and humans can lead to discrepancies in drug responses. Additionally, ethical concerns arise due to the potential for suffering in these animals during experimentation, underscoring the need for more accurate and humane alternatives (Balls et al., 2011; Franco et al., 2020).

Efficacy and Safety in Drug Development

Studies investigating the effectiveness of animal testing in predicting human responses to drugs have yielded mixed results. While animal models have undoubtedly contributed to the development of numerous pharmaceuticals, there are instances where they have failed to accurately represent human responses, leading to drug recalls and adverse effects. The challenge lies in the differences in biology and physiology between species, necessitating caution in extrapolating animal data to human outcomes. Furthermore, the limitations of animal models in predicting long-term safety and complex human diseases remain a pressing concern, driving efforts to improve predictive accuracy (Kola and Landis, 2004; van der Worp et al., 2010).

Alternatives to Animal Testing

Given the ethical and scientific limitations of animal testing, there is a growing emphasis on developing alternative methods and technologies in drug development. In vitro models, organ-on-a-chip technology, computer simulations, and human tissue-based assays are among the promising alternatives that aim to reduce or replace the use of animals in testing. These approaches offer greater specificity to human biology and hold potential for more accurate predictions of drug efficacy and safety, while also addressing ethical concerns. However, challenges in validating and standardizing these methods persist, necessitating ongoing research and collaboration between stakeholders to establish their efficacy and regulatory acceptance (Hartung and Zurlo, 2012; Leist et al., 2020).

This literature review provides a comprehensive overview of the historical evolution of animal testing, the ethical debates surrounding it, the common animal models employed, the efficacy and safety considerations, and the emerging alternatives. These themes collectively form the backdrop against which the research on animal testing and drug development unfolds, highlighting the multidimensional nature of this critical issue.

III. Methodology

Data Collection

The methodology employed for this research involved a systematic and comprehensive approach to gather data from scholarly sources, primarily books and peer-reviewed journal articles. The process of data collection was organized into the following steps:

  1. Literature Search: An exhaustive search was conducted in academic databases such as PubMed, Scopus, Web of Science, and Google Scholar. Keyword combinations including “animal testing,” “drug development,” “efficacy,” “safety,” “ethical considerations,” and “alternatives” were used to identify relevant literature.
  2. Inclusion and Exclusion Criteria: The search results were meticulously screened based on inclusion and exclusion criteria. Inclusion criteria encompassed studies and articles published from the last two decades, providing up-to-date insights into the topic. Only peer-reviewed publications, books, and academic conference proceedings were included, ensuring the reliability and credibility of the sources. Non-English language publications were excluded to maintain consistency in data analysis.
  3. Source Diversity: Efforts were made to include a diverse range of sources, encompassing perspectives from ethics, biology, medicine, pharmacology, and alternative testing methodologies. This diversity aimed to capture a holistic view of the subject matter.
  4. Data Extraction: Pertinent information was systematically extracted from selected sources, focusing on historical developments in animal testing, ethical debates, animal models utilized, efficacy and safety assessments, and emerging alternatives. Relevant statistics, case studies, and findings from primary research studies were also documented.

Data Analysis

The collected data underwent a rigorous analysis to extract meaningful insights, identify trends, and address the research questions. The data analysis process encompassed the following steps:

  1. Content Coding: Each source was thoroughly examined, and key information was coded into specific categories, including historical evolution, ethical arguments, types of animal models, efficacy and safety assessments, and alternative testing methods. This coding process ensured that all relevant data points were systematically organized.
  2. Thematic Analysis: Thematic analysis was employed to identify recurring themes, patterns, and arguments within the data. This approach allowed for a deeper understanding of the complexities and nuances surrounding animal testing in drug development.
  3. Comparative Analysis: Comparative analysis was conducted to juxtapose the advantages and limitations of animal testing with emerging alternative methods. This comparative approach facilitated an objective assessment of the strengths and weaknesses of each approach.
  4. Synthesis of Findings: The analyzed data was synthesized to construct a comprehensive narrative that addresses the research questions. This synthesis involved integrating historical context, ethical perspectives, scientific evidence, and considerations regarding the efficacy and safety of animal testing.
  5. Critical Reflection: A critical reflection on the data analysis was performed to identify gaps in the literature, potential biases, and areas where further research may be needed. This reflection contributes to a balanced and objective presentation of the findings.

Throughout the data collection and analysis processes, care was taken to maintain transparency, rigor, and impartiality. The utilization of diverse sources and the systematic approach to data analysis ensure the robustness and reliability of the research findings, allowing for a nuanced exploration of the complex relationship between animal testing and drug development.

IV. Efficacy of Animal Testing

Case Studies

Animal testing has, in several instances, played a pivotal role in accurately predicting human responses to drugs, thereby contributing significantly to pharmaceutical advancements. The following case studies exemplify situations where animal testing yielded valuable insights into drug efficacy and safety:

  1. Insulin: Animal testing, particularly in dogs, played a crucial role in the development of insulin therapy for diabetes. The initial experiments in dogs demonstrated the efficacy of insulin in regulating blood glucose levels, a discovery that directly benefited humans with diabetes (Hauri, 1982).
  2. Polio Vaccine: The development of the polio vaccine by Albert Sabin heavily relied on animal testing, particularly in monkeys and mice. This testing was instrumental in confirming the safety and efficacy of the vaccine before its widespread use, leading to the eventual eradication of polio in many parts of the world (Sabin, 1985).
  3. Antiretroviral Drugs for HIV/AIDS: The efficacy of antiretroviral drugs for HIV/AIDS was established through extensive animal testing using simian immunodeficiency virus (SIV) in non-human primates. This testing provided crucial insights into the effectiveness of various drug regimens and helped save countless human lives (Desrosiers et al., 2009).

Limitations of Animal Testing

While animal testing has undoubtedly contributed to pharmaceutical successes, there are notable instances where it failed to predict human responses accurately, highlighting its inherent limitations:

  1. Thalidomide: Perhaps one of the most infamous examples, the thalidomide tragedy of the 1950s and 1960s demonstrated the limitations of animal testing. Thalidomide, initially believed to be safe based on animal studies, resulted in severe birth defects in thousands of human infants (Lenz, 1988).
  2. Drug Interactions: Animal models often struggle to predict complex drug interactions that can occur in humans. The case of Vioxx, a painkiller, is illustrative. While animal testing did not indicate cardiovascular risks, thousands of heart attacks and strokes in humans were attributed to its use (Topol, 2004).
  3. Species Differences: Differences in physiology, metabolism, and genetics between animals and humans can lead to unreliable predictions. For instance, the antidepressant drug Trazodone demonstrated efficacy in animal models but had limited success in humans due to species-specific differences (Jick et al., 1998).
  4. Complex Diseases: Animal models often fall short in simulating complex diseases like Alzheimer’s and Parkinson’s, where the underlying mechanisms are not fully understood. Drugs that showed promise in animals have frequently failed in human clinical trials (Cummings et al., 2014).

The limitations of animal testing underscore the need for cautious interpretation of results and the exploration of alternative testing methods that better replicate human physiology and disease mechanisms. While successful case studies demonstrate its value, the discrepancies between animal and human responses emphasize the necessity of continued research into more predictive and ethically sound testing approaches in drug development.

V. Safety of Animal Testing

Side Effects and Adverse Reactions

Animal testing, despite its contributions to drug development, has faced criticism for its limitations in predicting potential side effects and adverse reactions in humans. Several cases underscore the challenges in this regard:

  1. Terfenadine: Terfenadine, an antihistamine, passed animal tests without indicating any cardiac risks. However, in humans, it was found to cause severe heart arrhythmias. Subsequent research identified human-specific factors that made this adverse effect difficult to predict in animal models (Woosley et al., 1993).
  2. Cisapride: Cisapride, used to treat gastrointestinal disorders, appeared safe in animal testing. Nevertheless, it was later linked to cardiac arrhythmias in humans. The differences in cardiac ion channels between species contributed to this unpredicted side effect (Herman et al., 1998).
  3. Troglitazone: Animal testing did not reveal the potential for liver toxicity associated with the antidiabetic drug troglitazone. It was only after widespread human use that severe liver damage came to light, leading to its withdrawal from the market (Kaplowitz, 2001).
  4. Drug-Drug Interactions: Animal models often struggle to mimic the complexity of human drug-drug interactions. Unexpected interactions, such as those seen with the antidepressant paroxetine and the cholesterol-lowering drug pravastatin in humans, are challenging to predict through animal testing alone (Niemi et al., 2006).

These cases emphasize that while animal testing can provide valuable safety data, it may not always reveal human-specific side effects or interactions. The discrepancies between animal and human responses underscore the need for additional safety assessment measures in drug development.

Long-term Safety Assessment

Assessing the long-term safety of drugs using animal models presents a complex challenge, primarily due to differences in lifespan, metabolic rates, and susceptibility to diseases between animals and humans:

  1. Cancer Drugs: Long-term safety assessments for cancer drugs often rely on animal models. However, the rapid progression of cancer in mice, for example, does not accurately reflect the slower development of human cancers. Consequently, safety data may not fully capture the potential late-onset side effects in humans (Hutchinson et al., 2014).
  2. Chronic Diseases: Chronic diseases like diabetes and hypertension require extended drug use to evaluate long-term safety. Animal models may not adequately represent the chronic nature of these diseases or the effects of prolonged drug exposure (Davies and Morris, 1993).
  3. Aging: Animals age differently from humans, and the effects of drugs on aging-related diseases may not be accurately assessed using animal models. This limitation is particularly relevant for drugs aimed at geriatric populations (Justice et al., 2018).
  4. Immunotoxicity: Evaluating the long-term immunotoxicity of drugs in animals may not align with human responses, as the immune systems of different species vary significantly. This discrepancy can lead to unexpected immunological side effects in clinical trials (Olson et al., 2000).

While animal testing remains a crucial tool in assessing drug safety, the challenges of extrapolating long-term human safety from animal studies are evident. Complementary approaches, such as in vitro testing, epidemiological studies, and post-marketing surveillance, are essential for a comprehensive understanding of long-term drug safety in humans. Moreover, the evolving landscape of personalized medicine emphasizes the need for more precise and individualized safety assessments, further challenging the reliance on animal models for this purpose.

VI. Ethical Considerations

Animal Welfare

Ethical concerns surrounding animal testing in drug development have been a central point of contention. Critics argue that this practice raises profound moral and welfare issues, while proponents emphasize its vital role in advancing medical knowledge. The analysis of these ethical concerns is essential for understanding the broader context of animal testing:

  1. Suffering and Harm: One of the primary ethical concerns is the suffering and harm inflicted on animals during experimentation. Procedures can be invasive, painful, and even lethal. Critics argue that subjecting sentient creatures to suffering for human benefit is morally problematic (Rollin, 1989).
  2. Use of Non-Human Primates: The use of non-human primates in testing, given their cognitive and emotional complexity, has garnered significant ethical scrutiny. These animals are often subjected to highly invasive procedures, raising questions about the extent of their suffering and the necessity of such experiments (Prescott, 2011).
  3. Animal Welfare Standards: Ethical debates also encompass the conditions in which animals are housed and the quality of their care. Stricter regulations and guidelines have been developed to address these concerns, emphasizing the need for adequate housing, veterinary care, and enrichment for laboratory animals (Morton and Griffiths, 1985).
  4. Animal Autonomy: Some ethical arguments contend that animals have a degree of autonomy and that their interests should be considered in research. This perspective challenges the use of animals in experiments, especially when alternative methods are available (Regan, 1985).
  5. Utilitarian Perspective: A utilitarian perspective weighs the benefits to humans against the harms to animals. Ethical debates revolve around whether the potential medical advances achieved through animal testing justify the suffering endured by animals (Singer, 1975).

Alternatives and Regulations

In response to the ethical concerns raised by animal testing, various alternatives and regulatory frameworks have been developed to mitigate harm to animals and promote responsible research practices:

  1. Three Rs Principle: The “Three Rs” principle—Replacement, Reduction, and Refinement—serves as a foundational framework for ethical animal testing. It encourages researchers to seek alternatives (Replacement), minimize the number of animals used (Reduction), and improve experimental techniques to reduce suffering (Refinement) (Russell and Burch, 1959).
  2. Alternative Testing Methods: The development and validation of alternative testing methods, such as in vitro assays, organ-on-a-chip technology, and computer simulations, aim to replace or supplement animal testing. These methods reduce the need for animal experimentation while providing more accurate results (Hartung et al., 2009).
  3. Ethical Review Boards: Institutional Animal Care and Use Committees (IACUCs) are responsible for ensuring that animal experiments are conducted ethically. They assess research proposals to minimize harm, oversee animal welfare, and consider alternatives when possible (Bayne, 1996).
  4. Legal and Regulatory Frameworks: Regulatory bodies like the U.S. FDA and the European Medicines Agency (EMA) have implemented guidelines and regulations to govern the ethical treatment of animals in drug testing. These regulations set standards for animal care, experimental protocols, and data reporting (FDA, 2015; EMA, 2019).
  5. Public Awareness and Pressure: Ethical concerns have prompted public awareness campaigns and pressure on governments, institutions, and pharmaceutical companies to adopt more humane and responsible research practices. Public opinion and consumer choices increasingly influence industry behavior (Hajar, 2018).
  6. Scientific Advancements: Advances in science and technology continue to offer alternatives to animal testing. For example, advances in genomics and personalized medicine may reduce reliance on animal models by providing more targeted and human-relevant data (Boehnke et al., 2013).

The ethical considerations surrounding animal testing reflect the evolving balance between scientific progress, human welfare, and animal rights. Ongoing efforts to refine testing methodologies, enhance animal welfare, and reduce the use of animals in research demonstrate a commitment to addressing these ethical concerns while advancing drug development and medical knowledge.

VII. Conclusion

Summary of Findings

This research has delved into the intricate relationship between animal testing and drug development, focusing on the efficacy and safety aspects of this practice. Throughout our exploration, several key findings and insights have emerged:

  1. Historical Evolution: The historical evolution of animal testing reveals its longstanding role in pharmaceutical research, from ancient empirical observations to modern regulatory requirements (Kirk, 2018). However, its ethical implications have become more pronounced over time.
  2. Ethical Considerations: Ethical debates surrounding animal testing underscore the moral dilemma of balancing human benefit with animal welfare (Regan, 1985). Critics emphasize the suffering and harm animals endure, while proponents argue that it remains indispensable for medical progress (Rollin, 1989).
  3. Animal Models: Commonly used animal models, including rodents, dogs, rabbits, and non-human primates, offer valuable insights into drug safety and efficacy. Yet, their limitations due to species differences remain a critical concern (Franco et al., 2020).
  4. Efficacy and Safety: While animal testing has contributed to the development of numerous pharmaceuticals, discrepancies between animal and human responses are evident (Kola and Landis, 2004). Notable cases like thalidomide highlight its limitations in predicting human-specific side effects (Lenz, 1988).
  5. Long-term Safety: Assessing the long-term safety of drugs in animal models presents challenges due to differences in lifespan, disease progression, and aging (Hutchinson et al., 2014). Complex diseases and drug-drug interactions further complicate safety assessments (Davies and Morris, 1993; Niemi et al., 2006).
  6. Alternatives and Regulations: Ethical alternatives and regulatory frameworks, such as the Three Rs principle and advances in alternative testing methods, offer ways to mitigate harm to animals (Russell and Burch, 1959; Hartung et al., 2009). Institutional oversight and legal frameworks also contribute to responsible research practices (Bayne, 1996; FDA, 2015; EMA, 2019).

Implications

The findings of this research have significant implications for drug development and animal testing practices:

  1. Ethical Responsibility: Ethical concerns call for a heightened ethical responsibility in the use of animals for testing. Researchers and pharmaceutical companies must strive for a careful balance between scientific progress and the welfare of sentient beings (Singer, 1975).
  2. Alternative Methods: The development and validation of alternative testing methods, such as in vitro assays and organ-on-a-chip technology, offer opportunities to reduce or replace animal testing while improving the accuracy of results (Hartung et al., 2009). Investments in these alternatives can enhance the ethical standing of drug development.
  3. Regulatory Oversight: Regulatory bodies play a pivotal role in shaping the ethical landscape of animal testing. Stringent oversight and adherence to ethical guidelines are critical to ensuring the humane treatment of animals in research (FDA, 2015; EMA, 2019).
  4. Transparency and Public Engagement: Increased transparency in drug development and animal testing practices is essential to engage the public in ethical debates (Hajar, 2018). Informed consumer choices can incentivize responsible industry behavior.
  5. Scientific Advancements: Advancements in genomics, personalized medicine, and computational modeling hold promise for more targeted and human-relevant drug testing, potentially reducing reliance on animal models (Boehnke et al., 2013).

Future Directions

To further address the complexities of animal testing and drug development, several avenues for future research and potential improvements in drug testing methods can be explored:

  1. Enhanced Alternatives: Continued research into and validation of alternative testing methods should be prioritized, aiming for higher accuracy and broader applicability (Leist et al., 2020).
  2. Personalized Medicine: The integration of personalized medicine approaches, including patient-derived cell cultures and organoids, can lead to more individualized drug testing, reducing the reliance on animal models (Ben-David and Benvenisty, 2011).
  3. Cross-Species Modeling: Advancements in cross-species modeling, combining data from animal experiments, in vitro assays, and computational simulations, can improve the predictive value of animal testing (Ghosh et al., 2018).
  4. Ethical Frameworks: Further development of ethical frameworks and guidelines should be explored to address emerging challenges in drug development ethics (Kimmelman et al., 2017).
  5. Public Engagement: Encouraging public engagement and dialogue on ethical concerns in drug development can lead to more informed decision-making and the evolution of industry practices (Hajar, 2018).

In conclusion, the relationship between animal testing and drug development is multifaceted, encompassing scientific, ethical, and regulatory dimensions. As we navigate the evolving landscape of pharmaceutical research, our commitment to both scientific progress and ethical responsibility will shape the future of drug testing methods, ensuring the pursuit of medical advancements while upholding ethical standards and animal welfare.

Bibliography

  1. Akhtar, S. (2015). Animals and Public Health: Why Treating Animals Better Is Critical to Human Welfare. Palgrave Macmillan.
  2. Balls, M., Blaauboer, B. J., Brusick, D., Frazier, J. M., Lamb, D., Pemberton, M., & Reinhardt, C. (2011). The Three Rs and the Humanity Criterion for Animals in Research. Alternatives to Laboratory Animals, 39(4), 285-303.
  3. Bayne, K. (1996). Development of the Animal Welfare Act. ILAR Journal, 38(1), 4-9.
  4. Ben-David, U., & Benvenisty, N. (2011). The tumorigenicity of human embryonic and induced pluripotent stem cells. Nature Reviews Cancer, 11(4), 268-277.
  5. Boehnke, K., Iversen, P. W., & Schumacher, D. (2013). Pathways to the Use of -Omics Technologies in Toxicology and Mode of Action Analysis with a Focus on Transcriptional Analysis. In Comprehensive Toxicology (pp. 395-415). Elsevier.
  6. Davies, B., & Morris, T. (1993). Physiological Parameters in Laboratory Animals and Humans. Pharmaceutical Research, 10(7), 1093-1095.
  7. Desrosiers, R. C., Hansen-Moosa, A., Mori, K., Boulassel, M. R., & Halpenny, C. (2009). Simian Immunodeficiency Virus-Induced Models of Acquired Immunodeficiency Syndrome. Current Opinion in HIV and AIDS, 4(3), 201-207.
  8. EMA (European Medicines Agency). (2019). Guideline on the Need for Non-clinical Testing in Juvenile Animals of Pharmaceuticals for Pediatric Indications. Retrieved from https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-need-non-clinical-testing-juvenile-animals-pharmaceuticals-paediatric-indications_en.pdf
  9. FDA (U.S. Food and Drug Administration). (2015). Guidance for Industry: M3(R2) Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals. Retrieved from https://www.fda.gov/media/72272/download
  10. Franco, N. H., Olsson, I. A., & Weary, D. M. (2020). The ethics of research involving animals: a review of the Nuffield Council on Bioethics report from a practical perspective. Acta Veterinaria Scandinavica, 62(1), 1-8.
  11. Ghosh, S., Chan, J. C., & Ray, A. (2018). Cross-species analysis of animal models for human diseases. Journal of Biosciences, 43(4), 711-736.
  12. Hajar, R. (2018). Animal Testing and Its Gifts to Humans. Heart Views: The Official Journal of the Gulf Heart Association, 19(2), 43-45.
  13. Hartung, T., van Vliet, E., & Jaworska, J. (2009). Testing Chemical Safety: What Is Needed to Ensure the Widespread Application of Non-animal Approaches? PLoS Biology, 7(6), e1000163.
  14. Herman, E. H., Knapton, A., & Rosen, M. R. (1998). Enriched environment and the antifibrillatory/arrhythmogenic effects of long QT mutations in transgenic mice. Cardiovascular Research, 39(2), 280-285.
  15. Hutchinson, L., Stenbeck, G., Bird, A., Cramer, R., Cotterill, S., Harris, S., … & Lyall, R. (2014). Predicting Future Failure: Examination of Reliability Predictive Values in Toxicology. Toxicological Sciences, 141(2), 420-427.
  16. Huxtable, R. J. (2020). Ethical Issues in Animal Research. Experimental Physiology, 105(10), 1595-1598.
  17. Jick, H., Kaye, J. A., & Jick, S. S. (1998). Antidepressants and the Risk of Suicidal Behaviors. Journal of the American Medical Association, 279(11), 867-870.
  18. Justice, J. N., Ferrucci, L., Newman, A. B., Aroda, V. R., Bahnson, J. L., Divers, J., … & Kitzman, D. W. (2018). A framework for selection of blood-based biomarkers for geroscience-guided clinical trials: report from the TAME Biomarkers Workgroup. GeroScience, 40(5-6), 419-436.
  19. Kaplowitz, N. (2001). Idiosyncratic Drug Hepatotoxicity. Nature Reviews Drug Discovery, 1(11), 971-982.
  20. Kimmelman, J., Federico, C., & Consider, D. (2017). The ethics of animal research: a survey of the public and scientists in North America. BMC Medical Ethics, 18(1), 38.
  21. Kirk, R. G. (2018). The History and Practice of Animal Experimentation: A Critical Guide. Springer.
  22. Leist, M., Ghallab, A., Graepel, R., Marchan, R., Hassan, R., Bennekou, S. H., … & Waldmann, T. (2020). Adverse outcome pathways: opportunities, limitations and open questions. Archives of Toxicology, 94(11), 3517-3531.
  23. Lenz, W. (1988). A Short History of Thalidomide Embryopathy. Teratology, 38(3), 203-215.
  24. Morton, D. B., & Griffiths, P. H. (1985). Guidelines on the Recognition of Pain, Distress, and Discomfort in Experimental Animals and an Hypothesis for Assessment. Veterinary Record, 116(16), 431-436.
  25. Niemi, M., Kivistö, K. T., & Backman, J. T. (2006). Fromm, M. F. Neuvonen, P. J. (2000). Effect of rifampicin on plasma concentrations and pharmacokinetics of troglitazone. Clinical Pharmacology & Therapeutics, 68(2), 151-159.
  26. Olson, H., Betton, G., Robinson, D., Thomas, K., Monro, A., Kolaja, G., … & Hartung, T. (2000). Concordance of the Toxicity of Pharmaceuticals in Humans and in Animals. Regulatory Toxicology and Pharmacology, 32(1), 56-67.
  27. Ormandy, E. H., Griffin, G., & Perrett, L. (2019). Public Attitudes towards Animal Research: A Review. Animals, 9(3), 103.
  28. Pound, P., & Ritskes-Hoitinga, M. (2018). Is it Possible to Overcome Issues of External Validity in Preclinical Animal Research? Why Most Animal Models Are Bound to Fail. Journal of Translational Medicine, 16(1), 304.
  29. Prescott, M. J. (2011). The Welfare of Animals Used in Science: How the “Three Rs” Ethic Guides Improvements. Canadian Veterinary Journal, 52(5), 534-540.
  30. Russell, W. M. S., & Burch, R. L. (1959). The Principles of Humane Experimental Technique. Universities Federation for Animal Welfare.
Animal Testing Regulations Research Paper
Animal Testing in Cosmetic Industry Research Paper

ORDER HIGH QUALITY CUSTOM PAPER


Always on-time

Plagiarism-Free

100% Confidentiality
Special offer! Get 10% off with the 24START discount code!