Content area
[...]prudent investments and measured risks at this eventually, a specific moment can raise the risk of success for a new Potential4,6. The different stages or phases of the drug development and discovery process are20-24 Drug Discovery Period25-31 * Initiate drug discovery program Combinatorial chemistry * Lead compound series identification * Additional compounds are made NCE’ s identified Drug Development and Registration Period32-35 * IND plan established and initiated * IND filed * Clinical studies initiated * NDA prepared and submitted * Drug launched into the market Drug Marketing and Line Expansion36-41 * Post-Marketing surveillance initiated * New clinical indications pursued * New dosage forms and formulations developed * Activities conducted to support marketing Drug Discovery and Development: Drug Discovery: New studies into a disease process that motivate scientists to create a new product to prevent or reverse the disease’s effects are the usual ways that researchers find new drugs42,44. * Numerous molecular compound tests to identify potential protective effects against a wide range of illnesses * Current therapies with unexpected side effects45–51 * New technologies, such as those that offer novel approaches to genetic material manipulation or the delivery of pharmaceuticals to particular bodily locations52–54. Drug Development: After discovering a compound that shows promise for development, scientists carry out tests in order to understand more about60–66: * Its distribution, metabolism, excretion, and absorption * Its possible advantages and modes of action * The ideal dosage and method of delivery * Adverse effects also known as toxicity * The ways in which it impacts different age groups (e.g., by gender, race, or ethnicity) differently * How it affects other medications and therapies * Its efficacy in comparison to comparable medications Preclinical Trials: ICH has established a basic guideline for the technical necessities of acceptable preclinical drug development. Prior to the start of a clinical trial, researchers review existing data about the drug to formulate research questions and objectives75. * Selection criteria for participants * Number of people take part of the study * Duration of study * Dose and route of administration of dosage form * Assessment of parameters * Data collection and analysis Phase 1:
INTRODUCTION:
Since ancient times, humanity has been engaged in a never-ending battle with illnesses. Even if numerous pharmacological substances have been discovered and modified to increase their dependability for use as a remedy, there are still numerous obstacles that prevent them from being effective enough to be deemed a “successful” discovery1. A new molecule is created or released onto the market for the good of society after a lengthy process known as drug discovery. In overall, the drug development process takes 12 to 15 years to produce a drug molecule completely developed into a novel medication candidate2.
We are able to guess on how long this process will take and how much money will need to be spent before it can be released onto the market. From the beginning of the drug discovery process to a possible cure, it costs more than $1 billion potential medication3. The process of drug discovery involves several phases of drug development, including initial hit identification, pre-clinical and clinical studies, FDA review, and post- marketing surveillance to monitor the Efficacy of the medication once a large population has been reached4.
In this Review, we will go over each step in detail to have a better understanding of thorough understanding of the domain and its present state. The intricate path a drug takes this entire procedure necessitated that the researchers go through advanced preclinical research, applications for investigational new drugs (INDs), and Clinical trials prior to a medication’s eventual approval for sale from the FDA5. Researchers may investigate into fresh perspectives on the development or progression of a disease, which may reveal a new pathway that a novel medication can target4. As an alternative, many businesses carry out a thorough trial procedure to determine the compound of interest. Actually, prudent investments and measured risks at this eventually, a specific moment can raise the risk of success for a new Potential4,6. These days, the drug discovery process goes all the way from Using computer simulation software to screen hits in silicon through hit detection, improvement and molecular testing to reduce adverse effects, until the creation of a novel medication candidate7. Drug discovery has advanced significantly in recent years, largely due to advancements in molecular biology, computer-aided drug design software, medicinal chemistry, and chemistry of organic matter1. Drug discoveries were previously the result of serendipity and a continuous trial-and-error experimental process that could eventually derive into an unknown substance, later coming to be known as a drug; the discovery of penicillin is one such example9,10. Molecular biological disciplines’ thorough understanding of disease pathophysiology also aided in the discovery and development process8. Drug discoveries only began to actively enter the pipeline in the 1980s.It takes much longer and requires much more complex clinical data to report safety and efficacy on human subjects11,12. After a complete medical evaluation, the ultimate goal of drug discovery is to quickly introduce a safer and more effective medication to the market13,14
Drug Discovery consist of 4 major steps, including few subdivision in each of them:
1. Pre discovery
2. Preclinical Trials
3. Clinical Trials
4. Post marketing surveillance15
Objectives16
* Recognize the investigational drug success rates by stages.
* Define Pre-clinical studies
* Define Investigational New Drug Application – Phase I, Phase II, Phase III studies
* Define New Drug Application
* Define Phase IV studies
Periods in Drug Discovery and Development Process:
The entire drug discovery and development process, including its introduction into the commercial market, is thought to take five to ten years and cost approximately $1.7 billion to complete successfully [17-19]. The different stages or phases of the drug development and discovery process are20-24
Drug Discovery Period25-31
* Initiate drug discovery program Combinatorial chemistry
* Lead compound series identification
* Additional compounds are made NCE’ s identified
Drug Development and Registration Period32-35
* IND plan established and initiated
* IND filed
* Clinical studies initiated
* NDA prepared and submitted
* Drug launched into the market
Drug Marketing and Line Expansion36-41
* Post-Marketing surveillance initiated
* New clinical indications pursued
* New dosage forms and formulations developed
* Activities conducted to support marketing
Drug Discovery and Development:
Drug Discovery:
New studies into a disease process that motivate scientists to create a new product to prevent or reverse the disease’s effects are the usual ways that researchers find new drugs42,44.
* Numerous molecular compound tests to identify potential protective effects against a wide range of illnesses
* Current therapies with unexpected side effects45–51
* New technologies, such as those that offer novel approaches to genetic material manipulation or the delivery of pharmaceuticals to particular bodily locations52–54. Thousands of compounds could be viable candidates for medical treatment development at this point. However, only a few compounds appear promising after preliminary testing and allow additional research55–59.
Drug Development:
After discovering a compound that shows promise for development, scientists carry out tests in order to understand more about60–66:
* Its distribution, metabolism, excretion, and absorption
* Its possible advantages and modes of action
* The ideal dosage and method of delivery
* Adverse effects also known as toxicity
* The ways in which it impacts different age groups (e.g., by gender, race, or ethnicity) differently
* How it affects other medications and therapies
* Its efficacy in comparison to comparable medications
Preclinical Trials:
ICH has established a basic guideline for the technical necessities of acceptable preclinical drug development. Preclinical research in the drug development process involves evaluating the drug’s safety and efficacy in animal species that conclude to prospective human outcome. Preclinical trials also need to obtain approval from the relevant regulatory authorities, who must ensure that trials are conducted in a safe and ethical manner and would only give approval for those drugs which are confirmed to be safe and effective67,68.
There are two methods for carrying out the pre-clinical trials: toxicology and general pharmacology. The pharmacokinetic and pharmacodynamic characteristics of drugs are the focus of pharmacology.
Investigating undesirable pharmacological effects in appropriate animal models and keeping an eye on them in toxicological research are crucial. Pharmacokinetic studies are essential for determining the parameters of safety and efficacy in terms of distribution, metabolism, excretion, and absorption. These studies provide data on the rate of absorption for various administration routes, which aids in the choice of dosage form, distribution, metabolism rate, and elimination rate—all of which influence the drug’s half-life. The drug’s half-life provides clarification on its safety profile, which is necessary for regulatory bodies to approve a medication. The drug’s therapeutic efficacy is explained by its distribution mechanism, which is dependent on both its affinity and bioavailability. The possibility of going through stages of the biotransformation process and the production of drug metabolites is provided by drug metabolism. It also aids in absorbing the enzymes and reactions that take place during biotransformation69.
Both in-vitro and in-vivo tests that assess the drug’s toxicological effects can be used to conduct toxicological studies. The direct effects on cell proliferation and phenotype can be examined through in-vitro investigations. Toxicological effects can be determined both qualitatively and quantitatively through in-vivo investigations. Choosing the right animal species for a toxicity study is crucial because many medications are species specific. Clinical studies frequently rely on in- vivo investigations to assess the product’s pharmacological and toxicological effects, including its mode of action70-72.
The Investigational New Drug Process (IND):
Before starting clinical research, drug developers must submit an Investigational New Drug application to the FDA73. In the IND application, developers must include:
* Preclinical and toxicity study data
* Drug manufacturing information
* Clinical research protocols for studies to be conducted
* Previous clinical research data (if any)
* Information about the investigator/ developer74
Clinical Trials:
Clinical trials are carried out in individuals (volunteers) and have the purpose to provide answers to specific questions regarding the safety and effectiveness of medications, vaccines, other therapies, or novel ways to use existing treatments. Clinical trials conform to a specific study protocol that is created by the researcher, investigator, or manufacturer. As the developers design the clinical study, they will think about what they want to accomplish for each of the various Clinical Research Phases and begin the Investigational New Drug Process (IND), a procedure they must go through prior to clinical research. Prior to the start of a clinical trial, researchers review existing data about the drug to formulate research questions and objectives75.
* Selection criteria for participants
* Number of people take part of the study
* Duration of study
* Dose and route of administration of dosage form
* Assessment of parameters
* Data collection and analysis
Phase 1: Safety and dosage:
Phase I trials are the initial drug tests that involve lesser healthy human participants. Phase 1 typically involves 20 to 80 healthy volunteers who have the illness or condition. Patients are typically only used when a drug’s mechanism of action suggests that healthy individuals will not tolerate it. However, researchers perform Phase 1 trials on patients with that type of diabetes if a new medication is suggested for use in those patients. Pharmacodynamics in the human body are the subject of carefully monitored phase 1 studies. To determine what dosage a drug can tolerate in the body and what are its acute side effects, researchers modify the dosage schedule based on data from animal studies. Researchers understand the research mechanism of action, the side effects that come with increasing the dosage, and information regarding effectiveness as a Phase 1 trial progresses. This is essential for Phase 2 study design. Nearly 70% of medications advance to the following stage76.
Phase 2: Efficacy and side effects:
Phase II trials are carried out on larger patient populations (few hundred) with the goal of maintaining the Phase I safety evaluations and assessing the drug’s effectiveness. To determine whether the medication will be therapeutic, these trials are insufficient. Phase 2 studies give researchers more information about safety. These data are used by researchers to create new Phase3 research protocols, improve research methods, and enhance their research questions. Approximately 33% of medications advance to the following stages76.
Phase 3: Efficacy and adverse drug reactions monitoring:
Phase 3 studies are planned by researchers to demonstrate whether or not a product benefits a particular group of people. These studies, which include 300–3,000 volunteers, are sometimes referred to as pivotal studies. The majority of the safety data is provided by phase 3 studies. It’s possible that the earlier study ignored less frequent side effects. However, because phase 3 studies involve a larger number of volunteers and last longer, the findings are more likely to identify rare or long- lasting side effects. About 25–30% of medications advance to the following stage of clinical trials76.
New Drug Application:
The complete story of a drug molecule is expressed in a New Drug Application (NDA). Its goal is to confirm that a medication is both safe and effective for the intended use in the subjects of the study. The NDA must contain all information about a drug, from preclinical data to Phase 3 trial data. Reports on all studies, data, and analyses must be included by developers77. In addition to the results of clinical trials, developers need to include:
* Proposed labeling
* Safety updates
* Drug abuse information
* Patent information
* Institutional review board compliance information
* Directions for use
FDA Review:
After receiving a completed NDA, the FDA review team may need six to ten months to decide whether to approve the document. The FDA review team will reject the NDA if it receives an incomplete NDA.
Working with the developer to update prescribing information is crucial if the FDA determines that a drug has been found to be safe and effective for its intended use. Labeling is the term for this. The label clearly outlines the guidelines for drug use and the basis for approval. However, before the medication can be authorized for sale, the remaining problems must be resolved. In other situations, the FDA requires more research. The developer can decide at this point whether or not to carry out additional development. There are procedures for filing an official appeal if a developer disagrees with an FDA decisions78.
Phase 4: Post-Market Drug Safety Monitoring:
After the FDA has approved a drug or device, phase 4 trials are carried out. These studies are also known as post-marketing surveillance, which includes ongoing technical support and pharmacovigilance following approval. Phase 4 trials employ a variety of observational techniques and assessment patterns to measure the safety, cost-effectiveness, and effectiveness of an intervention in real-world situations. Phase IV studies may be conducted by the sponsoring company for competitive or other reasons, or they may be required by regulatory bodies (e.g., labeling changes, risk management/minimization action plans). As a result, the real picture of a drug’s safety basically takes place over the course of the months and even years that make up its shelf life. The FDA can decide to add precautions to the dosage or practice information, as well as other events for more serious adverse drug reactions, after reviewing reports of complications with both prescription and over-the-counter medications79,80.
Software Used in Drug Discovery:
Since the release of modern technology and associated tools, the drug discovery process has undergone significant development. These resources have not only helped to save time and effort, but they have also enhanced research results81. By boosting productivity in both the analytical and medicinal domains, these tools have assisted the pharmaceutical industries in staying competitive and up to date with the market. The creation of ChemDraw, a drawing tool that enables chemists worldwide to depict a compound’s structure in a very simple and direct manner, is one such important development82,83. One of its many important features is the ability to quickly identify the IUPAC names of the unique structures, along with their physical and chemical characteristics, among other things. AutoDock, Schrodinger, Discovery Studio, DDD Plus, MapCheck, and others are additional examples of this type84–87. In addition to saving the researchers a great deal of time and effort, these applications have improved the reproducibility and accuracy of the findings. The skill of capturing an image in a remarkable and complicated way that satisfies publishing requirements is a crucial component of data analysis or interpretation. By improving the generated image’s quality and impact, the software can help us in achieving the same goal88,89. Some of the common software used in the drug discovery process is listed in Table 1. It is outside the responsibility of this article to create all of the drug-related software’s examples and lists.
Table 1: List of most commonly used software in drug discovery
Sr. No | Name of the software | Original Use | Other use |
1 | ChemOffice | To draw chemical structures | Physical, chemical properties of a compound |
2 | AutoDock | Structure based drug design | Virtual screening, molecular docking |
3 | Schrodinger | Molecular modelling, simulation, dynamics | Protein structure analysis |
4 | ArgusLab | Drug designing | Molecular docking |
5 | Design-Expert | Optimization and screening of experiments | Validation response evaluation of experiments |
6 | QSAR-co | To develop QSAR models | Screening and Prediction of developed models |
7 | GraphPad prism | Statistical analysis of data | Comparison of significance of data |
8 | PyMol | For compound visualization | Pharmacophore modeling |
9 | Gromacs | Molecular dynamics | Simulation of amino acids, proteins etc. |
10 | Unipept | Metaproteomics study | Peptide characterization and analysis |
11 | Accord | Chem informatics | Storage and analysis of chemical structures |
12 | Jaguar | Quantum chemistry and calculations | Interpretation of spectrophotometric results |
13 | Gold | Flexible docking | Covalent docking pose prediction |
14 | SwissADME | Pharmacokinetics | Physicochemical evaluation |
15 | Ethowatcher | Behavioral analysis of animals | Physiological changes in laboratory animals |
CONCLUSION:
New drugs are an important part of modern medicine with the emergence of diseases. A few decades ago, a disease such as peptic ulcers was an indication for major surgery. The advent of new pharmacologic treatments and introduction of novel medications have reduced the serious complications of peptic ulcer disease.
Similarly, thanks to many new antiviral medications with which the outlook for HIV- infected patients has improved. It is important that physicians understand the process of drug discovery and development. Understanding the process can promote innovation, help physicians assess new products, underline the importance of reporting adverse drug events and provide physicians with the information to educate patients about participating in a clinical trial.
REFERENCES:
1.SIS/draw, and chemsketch. J. Chem. Inf. Comput. Sci. 2004; 44: 1886–1890 ().
2.Vrushali G. Raut, et al. Influence of newly Synthesized Superdisintegrant on Dissolution Rate Enhancement of Carbomazepine using Liquid. solid Compact Technique. Asian Journal of Research in Pharmaceutical Sciences. 2022.
3.Kiriiri, G. K., Njogu, P. M. and Mwangi, A. N. Exploring different Approaches to improve the success of drug discovery and development Projects: a review. 2020; 9.
4.Deore, A. B., Dhumane, J. R., Wagh, R. and Sonawane, R. The Stages of Drug Discovery and Development Process. Asian J. Pharm. Res. Dev. 2019; 7: 62–67.
5.Svennebring, A. M. and Wikberg, J. E. S. Net present value approaches for drug discovery. Springerplus. 2013; 2:1–7.
6.Nicolaou, K. C. Advancing the Drug Discovery and Development Process. Angew.Chemie. 2014; 126; 9280–9292.
7.Mohs, R. C. and Greig, N. H. Drug discovery and development: Role of Basic biological research. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2017; 3: 651–657.
8.Zhong, X. and Moseley, G. B. Mission possible: Managing innovation In drug discovery. Nat. Biotechnol. 2007; 25: 945–946.
9.Berdigaliyev, N. and Aljofan, M. An overview of drug discovery and Development.Future Med. Chem. 2020; 12: 939–947.
10. Jackson, M., Marks, L., May, G. H. W. and Wilson, J. B. The genetic Basis of disease. Essays Biochem. 2018; 62: 643–723.
11. Ban, T. A. The role of serendipity in drug discovery. Dialogues Clin. Neurosci. 2006; 8: 335–344.
12. Weiser, J., Weiser, A. A., Shenkin, P. S. and Still, W. C. Neighbor-list Reduction: Optimization for computation of molecular van der Waal and solvent-accessible surface areas. J. Comput. Chem. 19, 797–808 (1998).
13. Holford, N., Ma, S. C. and Ploeger, B. A. Clinical trial simulation: A Review. Clin. Pharmacol. Ther. 88, 166–182 (2010).
14. McKee, A. E., Farrell, A. T., Pazdur, R. and Woodcock, J. The Role of The U.S. Food and Drug Administration Review Process: Clinical Trial Endpoints in Oncology. Oncologist 15, 13–18 (2010).
15. Clayden, J., Moran, W. J., Edwards, P. J. and Laplante, S. R. The Challenge of atropisomerism in drug discovery. Angew. Chemie – Int. Ed. 48, 6398–6401 (2009).
16. Zisowsky, J., Krause, A. and Dingemanse, J. Drug development for Pediatric populations: Regulatory aspects. Pharmaceutics 2, 364–388 (2010).
17. Balganesh, T., Kundu, T. K., Chakraborty, T. K. and Roy, S. Drug Discovery Research in India: Current State and Future Prospects. 724–726 (2014).
18. Russu WA. The Climate is Right to Accelerate New Drug Development for Neglected Diseases. Outlook Emerg Drugs. 2012; 1: 1.
19. Frohlich E. Alternatives to Animal Procedures in Drug Development. J Mol PharmOrg Process Res. 2016; 4: 132.
20. Cacabelos R. The Complexity of Alzheimer’s disease PharmacogenomicsandMetabolomics in Drug Development.Metabolomics. 2016; 6: e145.
21. Shastri PN. FDA Expedited Drug Development Programs. J Pharmacovigil. 2016; 4: e156.
22. Subudhi BB, et al. Updates in Drug Development Strategies against Peptic ulcer. JGastrointest Dig Syst. 2016; 6: 398.
23. Shimizu T and Nakagawa K. Novel Drug Development of the Next- Generation T790M Mutant Specific Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors for the Treatment of Advanced Non-Small Cell Lung Cancer. Biochem Anal Biochem. 2016; 5: 258.
24. Zhao Y. Towards Structural Based Drug Development for Ebola Virus Disease. J Chem Biol Ther. 2016; 1: e101.
25. Geert ER, et al. Selection Strategy of In Vivo Models for Ophthalmic Drug Development in Diabetic Retinopathy. J Mol Genet Med. 2016; 10: 202.
26. Nayeem N, et al. Gallic Acid: A Promising Lead Molecule for Drug Development. J App Pharm. 2016;8:213.
27. Emmanuel A, et al. Preliminary Identification of Lactate Dehydrogenase Inhibitors towards Anticancer Drug Development. J Develop Drugs. 2015; 4: 132.
28. Elhassa GO and Alfarouk KO. Stem Cell Therapy in Drug Discovery and Development. J Pharmacovigilance. 2015; 3: e140.
29. Melchior ACB, et al. Bromodomain as New Targets in Drug Discovery. BiochemPharmacol (Los Angel) 2015; 4: e176.
30. Goldberg JM and Panoff J. Pediatric Oncology Drug Development: A Case Reportand Pathways Forward. Pediat Therapeut. 2014; 4: 202.
31. Lu DY et al. Anticancer Drug Development, a Matter of Money or a Matter of Idea? Metabolomics. 2015; 5: e134.
32. Prado CM, et al. New Pharmacological Targets for Asthma Drug Development. J Allergy Ther. 2014; 5: 170.
33. Bourgailh J, et al. ViewLux™ Microplate Imager for Metabolite Profiling: Validation and Applications in Drug Development. J Anal Bioanal Tech. 2014; 5: 185.
34. Lu DY and Che JY. Rethink Of Diabetes Treatment and Drug Development. Cell Dev Biol. 2014; 3 :e125.
35. Papanastasopoulos P. Functional Imaging in Cancer Drug Development: A Mini- Review. J Med Diagn Meth; 2014; 3: 157.
36. Lee MKK and Dilq. Drug Development in Cell Culture: Crosstalk from the Industrial Prospects. J Bioequiv Availab. 2014; 6: 096-114.
37. Shengqian Wu, et al. Role of In-silico modeling in Drug Development for Inhalation Treatment. J Mol Pharm Org Process Res. 2013; 1: 106.
38. Jackson R. What can systems pharmacology contribute to drug development? Disease modelling as a predictive tool. BioDiscovery 2012; 4: 4.
39. Ceron-Carrasco JP, et al. Application of Computational Drug Discovery Techniques for Designing New Drugs Against Zika Virus. Drug Des. 2016; 5: e131
40. Ganesan A and Barakat K. Target fishing: a key to unlock the One-To- Many puzzle in drug discovery. J Pharma Care Health Sys. 2016; 3: e141.
41. Valayil JM. Activation of Microbial Silent Gene Clusters: Genomics Driven Drug Discovery Approaches. Biochem Anal Biochem. 2016; 5: 276.
42. Bueno J. Antimicrobial Adjuvants Drug Discovery, the Challenge of Avoid the Resistance and Recover the Susceptibility of Multidrug- Resistant Strains. J Microb Biochem Technol. 2016; 8: 169-176.
43. Melchior ACB, et al. Bromodomain as New Targets in Drug Discovery. Biochem Pharmacol (Los Angel). 2015;4:e176.
44. Ashwini R. Ghorpade, et al., “Review on vesicular Drug Delivery system: Aquaomes”, “Asian Journal of Research in Chemistry” 2024 March-April ,0974-4150
45. Agrawal P. Stem Cell Therapy in Drug Discovery and Development. J Pharmacovigilance. 2015; 3: e140.
46. Zambon S, et al. Development of a Rapid and Cost Effective Assay for the Screening of Reversible Cytochrome P450 Inhibition in Parallel with Cyp3a4 Metabolism-Dependent Inhibition Using Recombinant Proteins. Pharm Anal Acta. 2015; 6: 389.
47. Vrushali G Raut, et al, “A Review Mechanism of Role of superdisintogrants in the Development of Mouth Dissolving Tablets”, “International Journal; Pharma Tech Research” 2011, 2455-9563.
48. Xiong J. Multiple Targeting Routes of MicroRNA-22 Inform Cancer Drug Discovery. Gene Technol. 2015; 4: e114.
49. Sanaki T, et al. Improvements in the High-Performance Liquid Chromatography and Extraction Conditions for the Analysis of Oxidized Fatty Acids Using a Mixed-Mode Spin Column. Mod Chem appl. 2015; 3: 161.
50. Baig MH, et al. Drug Discovery and In Silico Techniques: A Mini- Review. Enz Eng. 2015; 4: 123.
51. Tyagi AK and Prasad S. Drug Discovery Inspired by Mother Nature for Cancer Therapy. Biochem Physiol. 2015;4:e128.
52. Dembitsky VM. Astonishing Diversity of Natural Peroxides as Potential Therapeutic Agents. J Mol Genet Med. 2015; 9: 163.
53. Den Haan HD, et al. Application of Modern Drug Discovery Techniques in the Context of Diabetes Mellitus and Atherosclerosis. Drug Des. 2015;4:e125.
54. Marjeta U. Chemo Proteomics, a Valuable Tool for Biomarker and Drug Discovery. Mol Biol. 20143:e117.
55. Nazem A. Alzheimer’s Disease Drug Discovery is Misled by Wrong Animal Models. J Gerontol Geriat Res. 2014;3:e127.
56. Bueno J. Biosensors in Antimicrobial Drug Discovery: Since Biology until Screening Platforms. J Microb Biochem Technol. 2014; S10: 002.
57. R.R Shinde, et al, “Pharmacosome as a vesicular Drug Delivery system”, “Asian Journal of Research in Pharmaceutical Science”, 2022 Oct-Dec, 2231-5659.
58. Helieh S. Oz. Toxoplasmosis, Pancreatitis, Obesity and Drug Discovery. Pancreat Disord Ther. 2014; 4: 138.
59. Agrawal P. Drug Discovery and Development: An Insight into Pharmacovigilance. J Pharmacovigilance. 2014;2:e120.
60. Li Y and Yang ST. Advances in Human Pluripotent Stem Cells for Regenerative Medicine and Drug Discovery. J Tissue Sci Eng. 2014; 5: e127.
61. Chang HW. Drug Discovery and Bioinformatics of Marine Natural Products. J Marine Sci Res Dev. 2013;4:e121.
62. Shivani Deshmukh, et al, “A Review on Diverging approaches to Fabricate Polymeric Nanoparticles”, “Asian Journal of Research in Pharmaceutical Science”, 2023 Oct-Dec, 2231-5659
63. Sanket Nikam, et al, “Pulsatile Delivery of Drug for a Range of Diseases”, “Asian Journal of Research in pharmaceutical Sciences”, 2022 Oct-Dec. 2231-5659.
64. Mishra NK and Shukla M. Application of Computational Proteomics and Lipidomics in Drug Discovery. J Theory Comput Sci. 2014; 1: 105.
65. Feng Y and Wang N. The New Generation of Drug Discovery and its Analytical Technologies. J Bioequiv Availab. 2013;5:e42.
66. Ravindran S, et al. Significance of Biotransformation in Drug Discovery and Development. J Biotechnol Biomaterial. 2012; S13: 005.
67. Gupta D. Pharmacogenomics in Drug Discovery and Development. J Develop Drugs. 2013; 2: e126.
68. Pradeep KV, et al. Importance of ADME and Bioanalysis in the Drug Discovery. J Bioequiv Availab. 2013; 5: e31.
69. Barile FA. Pri. Nciples of Toxicological Testing. CRC Press, USA, 2008.
70. Bhagyashri V. Aiwale, et al, “A Review on in situ Gel of Gastro Retentive Drug Delivery System”, “Asian Journal of Research in Pharmaceutical Sciences”, 2022 Oct- Dec ,2231-5659.
71. Friedman LM, Furberg CD, Demets DL. Fundamentals of clinical trials. 4th ed. New York: Springer Science and Business Media LLC; 2010.
72. Faqi AS. A comprehensive guide to toxicology in preclinical drug development. Waltham, MA: Elsevier; 2013.
73. Kaustubh Jagtap, et al, “Quality by Design (gbd) concept Review in Pharmaceutical”, “Asian Journal of Research in Chemistry” 2022 July-Aug, 0974-4150.
74. Bharatee Chaudhari, Kartika Daniel, “A Validated RP-HPLC Method for simultaneous Estimation of Tizanidine and Nimesulide in Bulk and Pharmaceutical and Technology”, 2020 Sep, 0974-360x.
75. Vogel HG. Drug Discovery and Evaluation 2nd edition. Springer, USA, 2002.
76. Karara AH, Edeki T, McLeod J, et al. PhRMA survey on the conduct Of first-in- human clinical trials under exploratory investigational new Drug applications. Journal of Clinical Pharmacology, 2010; 50:380- 391.
77. Fitzpatrick S. The clinical trial protocol. Buckinghamshire: Institute of Clinical Research; 2005.
78. Friedhoff L. New Drugs: An Insider‘s Guide to the FDA‘s New Drug Approval Process for Scientists, Investors and Patients. New York, NY: PSPG Publishing; 2009.
79. FDA (2003). New Drug Approval Reports. http://www.fda.gov/cder/rdmt/default.htm.
80. FDA, The FDA and the Drug Development Process: How the FDA Insures that drugs are safe and effective, FDA Fact sheet, 2002.
81. Adams CP, and Brantner VV. New Drug Development: Estimating Entry from human clinical trials. Bureau of Economics Federal Trade Commission. 2003.
82. Neha B. Waghmode, et al, “Review on Colon Targeted Drug Delivery System”, “Asian Journal of Research in Chemistry”, 2024 May-June ,0974-4150.
83. Sisodiya, D., Pandey, P. and Dashora, K. Drug Designing Softwares and Their Applications in New Drug Discover. J. Pharm. Res. 5, 124– 126 (2012).
84. Li, Z., Wan, H., Shi, Y. and Ouyang, P. Personal experience with four Kinds of chemical structure drawing software: Review on chemdraw, Chemwindow, I
85. Hopkinson, G. A. The Accord Component Software Approach. J. Chem. Inf. Comput. Sci. 37, 143–145 (1997).
86. Akram, W. and Garud, N. Design expert as a statistical tool for Optimization of 5- ASA-loaded biopolymer-based nanoparticles using Box Behnken factorial design. Futur. J. Pharm. Sci. 7, (2021).
87. Van Der Spoel, D. et al. GROMACS: Fast, flexible, and free. J. Comput. Chem. 26, 1701–1718 (2005).
88. Nurisso, A., Bravo, J., Carrupt, P. A. and Daina, A. Molecular docking Using the molecular lipophilicity potential as hydrophobic descriptor: Impact on GOLD docking performance. J. Chem. Inf. Model. 52, 1319–1327 (2012).
89. Yuan, S., Chan, H. C. S. and Hu, Z. Using PyMOL as a platform for Computational drug design. Wiley Interdiscip. Rev. Comput. Mol. Sci.7, 1–10 (2017).
90. Gurdeep Singh, R. et al. Unipept 4.0: Functional Analysis of Metaproteome Data.J. Proteome Res. 18, 606–615 (2019).
Credit: Kashish R. Mulani, B.P. Chaudhari, V. K. Redasani, Kalyani Gardi
Copyright 2025 A & V Publication, distributed by Contify.com