Explore chapters and articles related to this topic
Caenorhabditis elegans
Published in Iniewski Krzysztof, Integrated Microsystems, 2017
Pouya Rezai, Sangeena Salam, P. Ravi Selvaganapathy, Bhagwati P. Gupta
Drug discovery is the process of identifying drugs for human diseases. Typically, this involves screening for a large number of chemical compounds against certain targets in cells using in vitro and in vivo disease models [1]. In general, the process includes five stages: target identification and validation, lead screening, optimization, preclinical development, and clinical trials. A protein or biochemical pathway that plays a key role in the origin or progression of the disease is identified in the target identification and validation stage. The lead screening stage involves screening of a large number of chemical compounds against the biomolecular target protein or gene in order to identify candidates with potential therapeutic effect. This is followed by an optimization stage in which small chemical modifications of the initial lead compounds are made and screened to produce an optimal chemical species. The preclinical stage involves testing the candidates in various other animal models for efficacy as well as toxicity, and finally, in the clinical stage, testing is carried out in humans on a smaller scale. If successful, the drug is made commercially available in the market.
Chitosan: A Versatile Biomaterial for the 21st Century
Published in Shakeel Ahmed, Aisverya Soundararajan, Marine Polysaccharides, 2018
A. Shajahan, V. Kaviyarasan, V. Narayanan, S. Ignacimuthu
Drug discovery and development involve highly challenging, laborious and expensive processes. Most of the drugs in the clinical phase, however, fail to achieve favourable clinical outcomes because they do not have the ability to reach the target site of action. A significant amount of the administrated drug is distributed over the normal tissues or organs which are not involved in the pathological process, often leading to severe side effects. An effective approach to overcome this critical issue is the development of drug delivery systems which release the drugs or bioactive compounds. This could increase patient compliance and therapeutic efficacy of pharmaceutical agents through improved pharmacokinetics and biodistribution [176–178].
The Role of Chemoinformatics in Modern Drug Discovery and Development
Published in Devrim Balköse, Ana Cristina Faria Ribeiro, A. K. Haghi, Suresh C. Ameta, Tanmoy Chakraborty, Chemical Science and Engineering Technology, 2019
The area that has benefited the maximum form chemoinformatics is drug design and development. Various developments have been made in the area of lead discovery and optimization. It is a vital link between theoretical design and drug design in vitro. A huge investment and strong competition in the area of drug design may lead to curtailing the hit and trial approach. A theoretical and computational approach was targeted wherein the active molecules would be tailored accordingly making the drug design process efficient. The usual drug discovery and development process take around 10–15 years and cost approximate 1 billion USD. Figure 15.1 shows the schematics of a traditional drug discovery and development process.
Rediscovering the discovered: the new paradigm in repurposing drugs
Published in Indian Chemical Engineer, 2020
Togapur Pavan Kumar, Srivari Chandrasekhar, Prathama S. Mainkar
Fundamental science made an immense impact on humanity in the form of drugs and vaccines. Continuous transformation through technology gave birth to novel pharmacological and immunological innovations thereby delivering cost-effective and quality regimens to the public. One of the drivers for a robust healthcare system is drug discovery. A drug discovery programme is initiated if a clinical condition does not have suitable therapeutic options to support the treatment regime. Serendipity, repurposing and new inventions are popular processes to launch a new drug in the market. A new drug discovery project is time-consuming as it can spread between 12 and 15 years, which includes study of target, establishing a pharmacophore and lead molecule, clinical trials and final launch in the market. This process also involves huge investments, ∼ 1–2 billion US dollars, and can fail at the clinical trials or after the product reaches the market. Celecoxib, COX-2 inhibitor, faced resistance from the time of its launch in the market. For more than a decade, medical practitioners were hesitant to prescribe the drug for studies indicated the drug caused heart problems. The Food and Drug Administration (FDA), after evaluating a study result, stated the drug is safe than previously believed [10]. Vioxx, a pain reliever, in 2004 and Bextra, an arthritis drug, in 2005 were pulled from the market due to safety concerns [11]. These are a few incidences of drugs pulled back from market even after following all standard procedures. Therefore, in such cases discovery of a new drug may not be able to deliver returns in terms of treatment and investment.
Scheduling and control of high throughput screening systems with uncertainties and disturbances
Published in Production & Manufacturing Research, 2022
Adetola Oke, Laurent Hardouin, Xin Chen, Ying Shang
A successful drug discovery is an extremely time-consuming procedure, including initial target identification and validation, pre-clinical trials on animals, regulatory approval to start trials in humans, clinical trials, submission of marketing and manufacturing authorization, licensing review, product sale, and post-marketing surveillance (Major, 1998; Mayer et al., 2008; Mayr & Fuerst, 2008; Noah, 2010; Pereira & Williams, 2007). With the development of robotics and high-speed computing technology, it is feasible to develop automatic systems that can screen a large number of biochemical compounds in a short period. Such an automatic compound screening and analyzing process is called high-throughput screening (HTS) in drug discovery of pharmaceutical industries. HTS is a standard technology routinely employed in the pharmaceutical industry for drug discovery processes. It is used for initial screening in the process of drug discovery to reduce what is an almost infinite number of possible combinations of compounds to a reasonably few enough possibilities on which further testing can be carried out. Many HTS systems consist of several activities on several different resources. An HTS operation can incorporate multiple batches, each with hundreds of events, where a batch is a combination of all the operations to be performed on a set of substances for complete analysis. HTS provides a practical and efficient method to test a large number of synthetic compounds in miniaturized in vitro assays to identify hit targets of interest. Then, the chemical compounds that have therapeutic and useful pharmacological or biological activities, called leads, are evaluated and undergo lead optimization to identify promising lead compounds. Followed by the initial synthesis and animal testing in preclinical trials and three phases of clinical trials on humans, a drug can be put on the market after the Food and Drug Administration (FDA) approval.
Molecular modeling strategy to design novel anticancer agents against UACC-62 and UACC-257 melanoma cell lines
Published in Egyptian Journal of Basic and Applied Sciences, 2023
Abdullahi Bello Umar, Adamu Uzairu
In drug discovery research, the identification and validation of lead compounds and the determination of active binding sites of biological targets related to a particular lead compound performed through wet lab experiments are pretty expensive and time-consuming [10]. Computational approaches effectively reduce the time required to obtain valuable drugs and decrease their associated economic costs, making it possible to propose new potential drugs with low expenditures and selective targeting [11,12]. Quantitative structure–activity relationship (QSAR) modeling is an example of an in silico method, which can be used to understand drug action, design new compounds and screen chemical libraries [13,14]. QSAR studies have been used to identify important structural features responsible for the anticancer activity of drugs [15]. Quantitative structure–activity relationships are a significant factor in drug design; consequently, it is quite evident why many users of QSAR [16,17] are located in industrial research units. Combinatorial approaches are an influential tool in selection to speed up drug discovery, and with different mechanisms of action, this method is being adopted to cure cancer [1,18]. Molecular docking is an exceptional computational technique for screening a huge chemical library to detect prospective chemicals that could be used to discover the binding capability for a particular target. In the last two decades, molecular docking has become the model of structure-based virtual screening of several chemical databases [16, 19]. It is widely used to determine the proper orientation of drug molecules in the protein active site and their binding affinity. Previously, extensive molecular docking studies were conducted to explore the biological activity of many chemical materials [16,19]. It is widely used to determine the proper orientation of drug molecules in the protein active site and their binding affinity. Previously, extensive molecular docking studies were conducted to explore the biological activity of many chemical materials [19–21]