m
Our Mission Statement
This is Photoshop's version of Loremer Ipsn gravida nibh vel velit auctoregorie sam alquet.Aenean sollicitudin, lorem quis bibendum auci elit consequat ipsutis sem nibh id elit.
Follow Us
Top
Pharma Focus Asia – Transcell
7207
post-template-default,single,single-post,postid-7207,single-format-standard,mkd-core-1.0,highrise-ver-1.0,,mkd-smooth-page-transitions,mkd-ajax,mkd-grid-1300,mkd-blog-installed,mkd-header-vertical,mkd-sticky-header-on-scroll-up,mkd-default-mobile-header,mkd-sticky-up-mobile-header,mkd-dropdown-slide-from-bottom,mkd-dark-header,mkd-full-width-wide-menu,mkd-search-dropdown,mkd-side-menu-slide-from-right,wpb-js-composer js-comp-ver-5.0.1,vc_responsive

Blog

Pharma Focus Asia

News update on Oncologics for the month of September 2019

Published Date: September 2019
Location: Hyderabad, Telangana, India

Pre-clinical trials are the windows of clinical phases in drug discovery and developmental practice. Several invitro, invivo and insilico models have been put to use predicting safety and efficacy of drug candidates to qualify entering into clinical phase. Of these, there are some compelling pre-clinical models like that of human sourced stem cell based platform technologies with advantageous features for better prognosis., if adopted in total would revolutionise the very industry, spend, timelines and success.

human diseases and models of disease in practice, and have noted the failure of the latter to predict. Because of this, there has been a section advocating abandoning models and focusing on clinical trials in human patients; however, ethical hurdles to primary screening of molecules in humans are overwhelming. Medical research continues to remain dependent on model systems for establishing efficacy and safety prior to clinical applications. New approaches to validate cellular and animal models of disease that harmonise their behaviour with human disease are now being aggressively considered. These alternatives include reverse translation of human monogenetic disease to establish homologous cell-based disease models, the use of human sourced stem cells, and molecular fingerprinting of diseased tissues.

The early stage of discovery, sometimes known as pre-discovery, is the founding phase across both drug discovery and development. This stage sets the path to success if assumed honestly by the discovery team. Drug discovery Research & Development(R&D) is known to be a tedious and lengthy process which is predominantly staged and grouped as pre-clinical phase before reaching the consumer market place. Traditional discovery begins with ideation, identification of a target: a mechanism affected by a diseased condition followed by chemistry to meet the targeted needs of the affected mechanism. Proof of concept (PoC) is to demonstrate that the chemistry in discussion is promising and to further verify that the theory has practical potential. Concept and feasibility tests are performed in the early stages of the design and development process. These tasks are elaborate, expensive, and time intensive; however, performing them methodically and on the right, relevant model system(s) is crucial; it can help in mitigating risk to users and preventing the discovery of unexpected failures during verification and validation. By implementing a risk-based approach in the discovery and pre-clinical phase, the value can pay
off in revenue and avoiding potential disasters. Unexpected failures can lead to stopping the project, a complete redesign of the product and in worst case scenarios, shelving while reasons could be bad choice of pre-clinical models. Pre-discovery, discovery stages have the need to employ pre-clinical models while the robustness and relevance if is ruthless will lead to pre-clinical experimentation. There has been an age old belief that simple model systems provide a powerful tool for developing and exploring new therapeutics. But the world of human therapeutics is different from the world of other species as the two worlds make different assumptions while co-existing. Till date, the hypothesis that if we found drugs that ‘cured’ fly disease models and then successfully showed the same in the mouse model, the drug would magically enter clinical phase with all the approvals and regulations, and was assumed to be the winning recipe. Because it made sense theoretically, 95 per cent of failed trials did not change the deep rooted belief kicking aside the hard truth.


About www.transtoxbio.com products:

Why model systems at all? Before testing the drug within a human biological environment, conducting pre-clinical studies on model systems that yield preliminary efficacy, toxicity, pharmacokinetic, and safety information is the routine, reflecting on Why
models at all as the question. An increasing change in the drug development pipeline that has emerged in recent years has led to generation of different types of pre-clinical model systems that are surrogates for human disease. Traditionally, the process was limited to the study of drug efficacy in animal models, while the recent discovery and use of CRISPR/Cas9-mediated genome alteration has modernised our ability to generate pre-clinical models containing human pathogenic genetic variants. Any technology that holds great promise in increasing the correlation between pre-clinical and clinical disease prognosis and treatment translatability calls for compulsive adoption. There are broadly three pre-clinical model categories: in vitro (test tube or outside normal biological context), in vivo (within a biological entity like an animal), and in silico (computer simulation of the interactions). In vitro models involve testing a drug outside a living organism, normally on tissues or cells cultured in the laboratory. Initially, these models were built using 2-D systems using tissues or cell lines suspended in petri dishes. However, 3-D models have now been established, with spherical models (constructed using cell lines) and organoids (an artificially grown mass of cells that resembles a particular organ) becoming increasingly popular. The advantages of in vitro models lie in that they can be used easily for large-scale production in pharmaceutical companies. Additionally, the non-requirement of live animals decreases expenses and also dismisses the need for submission of animal protocols in accordance with the AWA (Animal Welfare Act). On a whole, in vitro models present far fewer ethical obligations than animal models do. That said, the absence of a live organism makes it difficult to predict how the drug being tested will interact with multiple organ systems. The lack of physiological exposure means that there is no complex, multicellular response that can be observed. It is also difficult to test the effect of larger, mechanical

human diseases and models of disease in practice, and have noted the failure of the latter to predict. Because of this, there has been a section advocating abandoning models and focusing on clinical trials in human patients; however, ethical hurdles to primary screening of molecules in humans are overwhelming. Medical research continues to remain dependent on model systems for establishing efficacy and safety prior to clinical applications. New approaches to validate cellular and animal models of disease that harmonise their behaviour with human disease are now being aggressively considered. These alternatives include reverse translation of human monogenetic disease to establish homologous cell-based disease models, the use of human sourced stem cells, and molecular fingerprinting of diseased tissues.

The early stage of discovery, sometimes known as pre-discovery, is the founding phase across both drug discovery and development. This stage sets the path to success if assumed honestly by the discovery team. Drug discovery Research & Development(R&D) is known to be a tedious and lengthy process which is predominantly staged and grouped as pre-clinical phase before reaching the consumer market place. Traditional discovery begins with ideation, identification of a target: a mechanism affected by a diseased condition followed by chemistry to meet the targeted needs of the affected mechanism. Proof of concept (PoC) is to demonstrate that the chemistry in discussion is promising and to further verify that the theory has practical potential. Concept and feasibility tests are performed in the early stages of the design and development process. These tasks are elaborate, expensive, and time intensive; however, performing them methodically and on the right, relevant model system(s) is crucial; it can help in mitigating risk to users and preventing the discovery of unexpected failures during verification and validation. By implementing a risk-based approach in the discovery and pre-clinical phase, the value can pay
off in revenue and avoiding potential disasters. Unexpected failures can lead to stopping the project, a complete redesign of the product and in worst case scenarios, shelving while reasons could be bad choice of pre-clinical models. Pre-discovery, discovery stages have the need to employ pre-clinical models while the robustness and relevance if is ruthless will lead to pre-clinical experimentation. There has been an age old belief that simple model systems provide a powerful tool for developing and exploring new therapeutics. But the world of human therapeutics is different from the world of other species as the two worlds make different assumptions while co-existing. Till date, the hypothesis that if we found drugs that ‘cured’ fly disease models and then successfully showed the same in the mouse model, the drug would magically enter clinical phase with all the approvals and regulations, and was assumed to be the winning recipe. Because it made sense theoretically, 95 per cent of failed trials did not change the deep rooted belief kicking aside the hard truth.

Why model systems at all? Before testing the drug within a human biological environment, conducting pre-clinical studies on model systems that yield preliminary efficacy, toxicity, pharmacokinetic, and safety information is the routine, reflecting on Why
models at all as the question. An increasing change in the drug development pipeline that has emerged in recent years has led to generation of different types of pre-clinical model systems that are surrogates for human disease. Traditionally, the process was limited to the study of drug efficacy in animal models, while the recent discovery and use of CRISPR/Cas9-mediated genome alteration has modernised our ability to generate pre-clinical models containing human pathogenic genetic variants. Any technology that holds great promise in increasing the correlation between pre-clinical and clinical disease prognosis and treatment translatability calls for compulsive adoption. There are broadly three pre-clinical model categories: in vitro (test tube or outside normal biological context), in vivo (within a biological entity like an animal), and in silico (computer simulation of the interactions). In vitro models involve testing a drug outside a living organism, normally on tissues or cells cultured in the laboratory. Initially, these models were built using 2-D systems using tissues or cell lines suspended in petri dishes. However, 3-D models have now been established, with spherical models (constructed using cell lines) and organoids (an artificially grown mass of cells that resembles a particular organ) becoming increasingly popular. The advantages of in vitro models lie in that they can be used easily for large-scale production in pharmaceutical companies. Additionally, the non-requirement of live animals decreases expenses and also dismisses the need for submission of animal protocols in accordance with the AWA (Animal Welfare Act). On a whole, in vitro models present far fewer ethical obligations than animal models do. That said, the absence of a live organism makes it difficult to predict how the drug being tested will interact with multiple organ systems. The lack of physiological exposure means that there is no complex, multicellular response that can be observed. It is also difficult to test the effect of larger, mechanical
Pre-clinical studies are devised to test the effect of the drug on the organism as a whole and also on a specific biological function or system.
CT01_Subadra_Article Final
CLINICAL TRIALS
26 PHARMA FOCUS ASIA ISSUE 36 – 2019
devices (as opposed to drugs) in an in vitro setting.

The High Throughput Screening (HTS) activity is part of the drug discovery process, and consists in selecting among thousands of molecules the ones that could have a pharmaceutical use in pre-clinical setting. To do HTS, large compounds libraries or toolbox of molecules are tested on a biological model showing a specific therapeutic target. Human stem cells and their progenies with self-renewable capacity, their ability to differentiate into several tissue progenitors, and their phenotypic responsive nature combined with suitability for Transcriptomics/proteomics define them as a good platform for screening to discover new potential drugs for human diseases. The ability to procure the source, harvest, culture in large scale, batch wise primary progenitors produce make adult stem cells based platforms as the best alternative tools in developing model systems with reproducible, reliable and relevant ones for pre-clinical adoption. These platforms complemented with robots, readers, and a good data mining system, make them a compelling option to replace the irrelevant animal and transformed cell line model systems. Beyond HTS, stem cell-based approaches, search for novel predictive biomarkers of developmental toxicity, and extend the experimental approach to other tissue specific cellular systems for the prediction of developmental neuro, osteo, hepatotoxicity. Well established differentiation protocols for certain adult cell types most susceptible to chemical
mediated toxicity, any chromosomal abnormality during early development like cardiac, bone, and neural cells have been successfully developed. For neural cell development, stem cells can be efficiently differentiated in vitro into cell types present in the nervous system like mature neurons, astrocytes, andoligodendrocytes, while the process of differentiation acts as a test platform to evaluate neurogenesis, neurotoxicity kind of end points. On the basis of this approach, new rapid and predictive in vitro screens for developmental neurotoxicity testing have been developed. Even in stem cell based model systems, there are two fundamental criteria like potency and unlimited source availability that make them suitable for investigations spanning traditional discovery to regulatory testings. New exciting avenues of research on the role of microRNA (miRNA) in toxicogenomics and the possibility of epigenetic effects on gene expression opens the possibility to discover new molecular endpoints that might contribute to a further understanding of chemical-mediated developmental toxicity on stem cells based model systems. Furthermore, in line with new directions for toxicity testing, in the light of advances in understanding biological responses to chemical stressors involving the mapping of toxicity pathways in differentiating human stem cells and identification of critical pathway perturbations that represent molecular initiation

No Comments

Leave a Comment