Toward a Personalized Use of Paclitaxel

Total Page:16

File Type:pdf, Size:1020Kb

Toward a Personalized Use of Paclitaxel Send Orders for Reprints to [email protected] 296 Recent Patents on Anti-Cancer Drug Discovery, 2019, 14, 296-297 COMMENTARY Toward a Personalized Use of Paclitaxel Gian L. Russo* National Research Council, Institute of Food Sciences, Avellino 83100, Italy When Wall and Wani presented in 1967, for the first time and its cause-effect relationship with toxicity and therapeutic at the 153rd National Meeting of the American Chemical efficacy in 295 patients affected by different solid tumors, Society, their preliminary data on the structure of a new namely ovarian, esophageal, breast cancers, and NSCLC compound, that they called Taxol, possessing novel anti- under three different chemotherapeutic regimens with PTX leukemic and antitumor agent [1], they probably did not pre- representing the key consistent agent. TC > 0.05 indicates the dict that in the following 50 years, Taxol would become the time at which the plasma concentration of PTX exceeds largest cancer drug ever manufactured, with an annual sale 0.05µM. Previous works from the same group demonstrated that peaked in 2000, reaching $1.6 billion (source that this parameter was associated with chemotherapeutic https://dtp.cancer.gov). Taxol was originally isolated from efficacy or severe toxicity in ovarian cancer patients treated the bark of the Pacific yew tree Taxus brevifolia and its with PTX plus carboplatin depending on the threshold con- clinical success became soon the emblem of naturally- centration of PTX exposure [14]. Chen and Zhang identified derived anticancer drugs. The structure was published on three ranges of TC > 0.05 distribution (26 to 30h; below 26h May 5, 1971 [2]. Following an agreement signed in 1991 and above 30h) and correlated them to the efficacy of the between NCI and Bristol-Myers Squibb (BMS) for the isola- chemotherapeutic treatment. In fact, they showed that in tion and formulation into drug of taxol from the bark of the 65.6% of PTX, TC > 0.05 in CR (Complete Remission) and PR Pacific yew tree [3], BMS changed the original name from (Partial Remission) were in the range of 26-30h, suggesting “Taxol” to “Paclitaxel” (PTX) and the drug was sold under that monitoring the PTX TC > 0.05, other than simply the blood the trademark name in US of “Taxol®” [3]. Foreign brand concentration of the agent, is essential to optimize individual names are: Anzatax, Asotax, Bristaxol, Praxel and Taxol treatment in different forms of solid cancers [13]. In the sec- Konzentrat. Paclitaxel has been approved to be used alone or ond part of their work, the authors focused on the correlation in combination with other drugs against solid tumors, such as between PTX TC > 0.05 and several indicators of drug toxicity, AIDS-related Kaposi Sarcoma (KS), breast cancer, Non- namely: leukopenia and leukopenic fever; neutropenia and Small Cell Lung Cancer (NSCLC) and ovarian cancer [4, 5], neutropenic fever; anemia and peripheral neurotoxicity. In but it is also being studied for the treatment of other types of all cases, they demonstrated that the higher the PTX TC > 0.05 (> 30h), higher were the degree of all forms of adverse ef- cancer, including esophageal cancer [6], pancreas [7], blad- fects in patients with ovarian cancer, esophageal cancer, der [8], prostate [9], and melanoma [10]. From a molecular breast cancer and NSCLC (Tables 3-6 in [13]). point of view, the pivotal work conducted in the Horwitz’ laboratory demonstrated in 1979 that PTX induces cell cycle The monopoly of PTX practically ended in the first dec- arrest in G2/M Phase promoting microtubule assembly [11]. ade of this millennium with the appearance on the market in As a result, microtubules are resistant to depolymerization 2005 of an innovative formulation of this drug marketed and their dynamicity is inhibited by blocking fundamental under the trade name of Abraxane® and representing a pro- functions in eukaryotic cells, such as mitosis. In cancer cells, tein-bound, injectable formulation of PTX, also known as this mechanism explains the antiproliferative effects associ- nanoparticle albumin-bound paclitaxel (nab-paclitaxel). The ® ated with PTX [11]. In later works, PTX analogues bearing clinical efficacy of Abraxane is significantly higher than photoreactive groups were used to identify the binding sites PTX in terms of response rate, increased time to disease pro- on distinct β-tubulin isotypes [12]. gression, increased survival and reduced toxicity [15]. Abraxane® has been FDA approved to be used alone or with Despite the fact that, surfing PubMed, the term “pacli- other drugs to treat: recurred or metastasized breast cancer, taxel” appears in title/abstract > 26,000 times, this compound advanced or metastasized non-small cell lung cancer (in and its clinical applications are still intensively studied. A combination with carboplatin) and metastasized pancreatic good example comes from the recent article authored by cancer (in combination with gemcitabine hydrochloride) [16- Chen and Zhang groups [13]. Here, the authors investigated 18]. As clearly reported by Sofias et al. (Fig. 2 in [19]), the ® the association of the pharmacokinetic parameter TC > 0.05 Abraxane projected sales by 2020 can raise $1.9 billion, while the prediction for Taxol® is lowering below $100 mil- *Address correspondence to this author at the Consiglio Nazionale delle lion. Based on this evidence, the work by Chen and Zhang Ricerche - Istituto Scienze dell'Alimentazione, via Roma, 64 - 83110 - Avel- [13] appears extremely timing since it inspires the extension lino, Italy; Tel: +39 0825 299331; E-mail: [email protected] of their study to compare the TC > 0.05 parameter between 2212-3970/19 $58.00+.00 © 2019 Bentham Science Publishers Toward a Personalized Use of Paclitaxel Recent Patents on Anti-Cancer Drug Discovery, 2019, Vol. 14, No. 4 297 Abraxane® and PTX. In addition, the application of this [10] Leon-Ferre RA, Markovic SN. Nab-paclitaxel in patients with method can be extended to test the efficacy of the growing metastatic melanoma. Expert Rev Anticancer Ther 2015; 15(12): 1371-7. family of other taxane anticancer agents approved by FDA in [11] Schiff PB, Fant J, Horwitz SB. Promotion of microtubule assembly monotherapy or in combined chemotherapeutic formulations in vitro by taxol. Nature 1979; 277(5698): 665-7. [20]. [12] Yang CH, Horwitz SB. Taxol: The first microtubule stabilizing agent. Int J Mol Sci 2017; 18(8): 1733-41. [13] Xin DS, Zhou L, Li CZ, Zhang SQ, Huang HQ, Qiu GD, et al. REFERENCES TC > 0.05 as a pharmacokinetic parameter of paclitaxel for therapeutic [1] Wall ME, Wani MC. 153rd National Meeting of the American efficacy and toxicity in cancer patients. Recent Pat Anticancer Chemical Society; Miami Beach (FL)1967. pp. M-006. Drug Discov 2018; 13(3): 341-7. [2] Wani MC, Taylor HL, Wall ME, Coggon P, McPhail AT. Plant [14] Zhang S, Sun M, Yuan Y, Wang M, She Y, Zhou L, et al. Correla- antitumor agents. VI. The isolation and structure of taxol, a novel tion between paclitaxel TC > 0.05 and its therapeutic efficacy and se- antileukemic and antitumor agent from Taxus brevifolia. J Am vere toxicities in ovarian cancer patients. Cancer Transl Med 2016; Chem Soc 1971; 93(9): 2325-7. 2(5): 131-6. [3] Martin V. Overview of paclitaxel (TAXOL). Semin Oncol Nurs [15] Gradishar WJ. Albumin-bound paclitaxel: A next-generation tax- 1993; 9(4 Suppl 2): 2-5. ane. Expert Opin Pharmacother 2006; 7(8): 1041-53. [4] Mekhail TM, Markman M. Paclitaxel in cancer therapy. Expert [16] Drugs.com. FDA approves abraxane 2005 [Available from: http:// Opin Pharmacother 2002; 3(6): 755-66. www.drugs.com/newdrugs/abraxaneamerican-pharmaceutical- [5] Goodman J, Walsh V. The Story of Taxol: Nature and Politics in partners-inc-american-bioscience-inc-metastaticbreast-cancer- the Pursuit of an Anti-Cancer Drug. 1st ed. Cambridge University 143.html]. Press: New York 2001; 323(7304): 115-25. [17] ClinicalTrials.gov. Albumin-bound paclitaxel (ABI-007) for pa- [6] Van Hagen P, Hulshof MC, Van Lanschot JJ, Steyerberg EW, Van tientswith advanced non-small cell lung cancer 2013 [Available Berge Henegouwen MI, Wijnhoven BP, et al. Preoperative Chemo- from: https://clinicaltrials.gov/ct2/show/study/NCT00540514]. radiotherapy for esophageal or junctional cancer. N Engl J Med [18] ClinicalTrials.gov. Phase III study of ABI-007(Albumin-bound 2012; 366(22): 2074-84. paclitaxel) plus gemcitabine versus gemcitabine in metastatic ade- [7] Ma WW, Hidalgo M. The winning formulation: The development nocarcinoma of the pancreas 2016 [Available from: https://clinical of paclitaxel in pancreatic cancer. Clin Cancer Res 2013; 19(20): trials.gov/ct2/show/NCT00844649]. 5572-9. [19] Sofias AM, Dunne M, Storm G, Allen C. The battle of "nano" [8] Vaughn DJ. Paclitaxel and carboplatin in bladder cancer: Recent paclitaxel. Adv Drug Deliv Rev 2017; 122: 20-30. developments. Eur J Cancer 2000; 36(Suppl 2): 7-12. [20] Ojima I, Lichtenthal B, Lee S, Wang C, Wang X. Taxane antican- [9] Obasaju C, Hudes GR. Paclitaxel and docetaxel in prostate cancer. cer agents: A patent perspective. Expert Opin Ther Pat 2016; 26(1): Hematol Oncol Clin North Am 2001; 15(3): 525-45. 1-20. .
Recommended publications
  • Computational Approaches for Drug Design and Discovery: an Overview
    Review Article Computational Approaches for Drug Design and Discovery: An Overview Baldi A College of Pharmacy, Dr. Shri R.M.S. Institute of Science & Technology, Bhanpura, Dist. Mandsaur (M.P.), India ARTICLE INFO ABSTRACT Article history: The process of drug discovery is very complex and requires an interdisciplinary effort to design Received 12 July 2009 effective and commercially feasible drugs. The objective of drug design is to find a chemical Accepted 21 July 2009 compound that can fit to a specific cavity on a protein target both geometrically and chemically. Available online 04 February 2010 After passing the animal tests and human clinical trials, this compound becomes a drug available Keywords: to patients. The conventional drug design methods include random screening of chemicals Computer-aided drug design found in nature or synthesized in laboratories. The problems with this method are long design Combinatorial chemistry cycle and high cost. Modern approach including structure-based drug design with the help Drug of informatic technologies and computational methods has speeded up the drug discovery Structure-based drug deign process in an efficient manner. Remarkable progress has been made during the past five years in almost all the areas concerned with drug design and discovery. An improved generation of softwares with easy operation and superior computational tools to generate chemically stable and worthy compounds with refinement capability has been developed. These tools can tap into cheminformation to shorten the cycle of drug discovery, and thus make drug discovery more cost-effective. A complete overview of drug discovery process with comparison of conventional approaches of drug discovery is discussed here.
    [Show full text]
  • Opportunities and Challenges in Phenotypic Drug Discovery: an Industry Perspective
    PERSPECTIVES Nevertheless, there are still challenges in OPINION prospectively understanding the key success factors for modern PDD and how maximal Opportunities and challenges in value can be obtained. Articles published after the analysis by Swinney and Anthony have re-examined the contribution of PDD phenotypic drug discovery: an to new drug discovery6,7 and have refined the conditions for its successful application8. industry perspective Importantly, it is apparent on closer examination that the classification of drugs John G. Moffat, Fabien Vincent, Jonathan A. Lee, Jörg Eder and as ‘phenotypically discovered’ is somewhat Marco Prunotto inconsistent6,7 and that, in fact, the majority of successful drug discovery programmes Abstract | Phenotypic drug discovery (PDD) approaches do not rely on knowledge combine target knowledge and functional of the identity of a specific drug target or a hypothesis about its role in disease, in cellular assays to identify drug candidates contrast to the target-based strategies that have been widely used in the with the most advantageous molecular pharmaceutical industry in the past three decades. However, in recent years, there mechanism of action (MoA). Although there is clear evidence that phenotypic has been a resurgence in interest in PDD approaches based on their potential to screening can be an attractive proposition address the incompletely understood complexity of diseases and their promise for efficiently identifying functionally of delivering first-in-class drugs, as well as major advances in the tools for active hits that lead to first-in-class drugs, cell-based phenotypic screening. Nevertheless, PDD approaches also have the gap between a screening hit and an considerable challenges, such as hit validation and target deconvolution.
    [Show full text]
  • Medicinal Chemistry for Drug Discovery | Charles River
    Summary Medicinal chemistry is an integral part of bringing a drug through development. Our medicinal chemistry approach enables clients to benefit from efficient navigation of the early drug discovery process through to delivery of preclinical candidates. DISCOVERY Click to learn more Medicinal Chemistry for Drug Discovery Medicinal Chemistry A Proven Track Record in Drug Discovery Services: Our medicinal chemistry team has experience in progressing small molecule drug discovery programs across a broad range • Target identification of therapeutic areas and gene families. Our scientists are skilled in the design and synthesis of novel pharmacologically active - Capture Compound® mass compounds and understand the challenges facing modern drug discovery. Together, they are cited as inventors on over spectrometry (CCMS) 350 patents and have identified 80 preclinical candidates for client organizations across a variety of therapeutic areas. As • Hit-finding strategies project leaders, our chemists are fundamental in driving the program strategy and have consistently empowered our clients’ - Optimizing high-throughput success. A high proportion of candidates regularly progress to the clinic, and our first co-invented drug, Belinostat, received screening (HTS) hits marketing approval in 2015. As an organization, Charles River has worked on 85% of the therapies approved in 2018. • Hit-to-lead We have a deep understanding of the factors that drive medicinal chemistry design: structure-activity relationship (SAR), • Lead optimization biology, physical chemistry, drug metabolism and pharmacokinetics (DMPK), pharmacokinetic/pharmacodynamic (PK/PD) • Patent strategy modelling, and in vivo efficacy. Charles River scientists are skilled in structure-based and ligand-based design approaches • Preparation for IND filing utilizing our in-house computer-aided drug design (CADD) expertise.
    [Show full text]
  • Overcoming the Immunosuppressive Tumor Microenvironment in Multiple Myeloma
    cancers Review Overcoming the Immunosuppressive Tumor Microenvironment in Multiple Myeloma Fatih M. Uckun 1,2,3 1 Norris Comprehensive Cancer Center and Childrens Center for Cancer and Blood Diseases, University of Southern California Keck School of Medicine (USC KSOM), Los Angeles, CA 90027, USA; [email protected] 2 Department of Developmental Therapeutics, Immunology, and Integrative Medicine, Drug Discovery Institute, Ares Pharmaceuticals, St. Paul, MN 55110, USA 3 Reven Pharmaceuticals, Translational Oncology Program, Golden, CO 80401, USA Simple Summary: This article provides a comprehensive review of new and emerging treatment strategies against multiple myeloma that employ precision medicines and/or drugs capable of improving the ability of the immune system to prevent or slow down the progression of multiple myeloma. These rationally designed new treatment methods have the potential to change the therapeutic landscape in multiple myeloma and improve the long-term survival outcome. Abstract: SeverFigurel cellular elements of the bone marrow (BM) microenvironment in multiple myeloma (MM) patients contribute to the immune evasion, proliferation, and drug resistance of MM cells, including myeloid-derived suppressor cells (MDSCs), tumor-associated M2-like, “alter- natively activated” macrophages, CD38+ regulatory B-cells (Bregs), and regulatory T-cells (Tregs). These immunosuppressive elements in bidirectional and multi-directional crosstalk with each other inhibit both memory and cytotoxic effector T-cell populations as well as natural killer (NK) cells. Immunomodulatory imide drugs (IMiDs), protease inhibitors (PI), monoclonal antibodies (MoAb), Citation: Uckun, F.M. Overcoming the Immunosuppressive Tumor adoptive T-cell/NK cell therapy, and inhibitors of anti-apoptotic signaling pathways have emerged as Microenvironment in Multiple promising therapeutic platforms that can be employed in various combinations as part of a rationally Myeloma.
    [Show full text]
  • Drug Discovery - Yesterday and Tomorrow: the Common Approaches in Drug Design and Cancer
    Cell & Cellular Life Sciences Journal Drug Discovery - Yesterday and Tomorrow: The Common Approaches in Drug Design and Cancer 1,2 1 3 Hamad ON *, Amran SIB and Sabbah AM Mini Review 1Faculty of Bioscience & Medical Engineering, Malaysia Volume 3 Issue 1 2University of Wasit, College of Medicine, Iraq Received Date: February 27, 2018 3Forensic DNA for research and training Centre, Al Nahrain University, Iraq Published Date: April 03, 2018 *Corresponding author: Oras Naji Hamad, Faculty of Bioscience & Medical Engineering, University of Technology, Malaysia, Tel: 01121715960; E-mail: [email protected] Abstract The process of drug discovery has undergone radical changes and development over years. Traditionally, the drugs were discovered by employing chemistry and pharmacology-based cautious approach. When natural products were the most important source of drugs or drug precursors, but the conventional randomized drug research phenomenon was no longer effective at that time due to many negatives of these approaches like: high expenses of discovering new drugs, time-consuming and reduced success guarantee. Thus, with the development of the era, the concept of “Rational Drug Design” has enabled drug target identification and validation to be more specific. In addition, several novel technologies and approaches have been introducing economics, proteomics and other omics areas such as 3D QSAR, pharmacophore modeling and other, which playing a promising role in accelerating the pace of drug discovery process. Their view of the current
    [Show full text]
  • The Impact of Early ADME Profiling on Drug Discovery and Development Strategy
    ADME Profiling The impact of early ADME profiling on drug discovery and development strategy The increased costs in the discovery and development of new drugs, due in part to the high attrition rate of drug candidates in development, has led to a new strategy to introduce early, parallel evaluation of efficacy and biopharmaceutical properties of drug candidates. Investigation of terminated projects revealed that the primary cause for drug failure in the development phase was the poor pharmacokinetic and ADMET (Absorption, Distribution, Metabolism, Discretion and Toxicity) properties rather than unsatisfactory efficacy. In addition, the applications of parallel synthesis and combinatory chemistry to expedite lead finding and lead optimisation processes has shifted the chemical libraries towards poorer biopharmaceutical properties. Establishments of high throughput and fast ADMET profiling assays allow for the prioritisation of leads or drug candidates by their biopharmaceutical properties in parallel with optimisation of their efficacy at early discovery phases.This is expected to not only improve the overall quality of drug candidates and therefore the probability of their success, but also shorten the drug discovery and development process. In this article, we review the early ADME profiling approach, their timing in relation to the entire drug discovery and development process and the latest technologies of the selected assays will be reviewed. iscovery and development of a new drug is a lead finding/selection process, which in general takes By Dr Jianling Wang long, labour-demanding process. Recent a minimum of two more years. Typically, the whole and Dr Laszlo Urban Dstudies1 revealed that the average time to process is fragmented into ‘Discovery’, ‘Development’ discover, develop and approve a new drug in the and ‘Registration’ phases.
    [Show full text]
  • Setting Fair Prices for Life-Saving Drugs by Bruce A
    Virtual Mentor American Medical Association Journal of Ethics January 2007, Volume 9, Number 1: 38-43. Policy forum Setting fair prices for life-saving drugs by Bruce A. Chabner, MD, and Thomas G. Roberts Jr., MD, MSocSci Cancer drugs are big business. Worldwide sales are projected to reach $25 billion in 2006 and to increase to almost $50 billion by 2010 [1]. This represents a startling growth in a segment of the drug industry once shunned by major pharmaceutical manufacturers as too high-risk and unprofitable. While a few drug companies, notably Bristol-Myers Squibb (BMS) and Pharmatalia, made significant profits on cancer drugs between 1970 and 1990 when the first effective combination therapies came into common practice, the turning point in this industrial segment occurred in 1992 with the approval of Bristol-Myers Squibb’s paclitaxel, which became a multibillion-dollar-per-year product by 1998. To understand our current concerns with cancer drug costs and their potential effect on medical care financing and access, one needs to be familiar with the paclitaxel experience. The story of paclitaxel’s discovery and commercial development reflects both the lack of interest that industry had in cancer drugs at that time and the sudden emergence of drug cost as a social justice issue. In 1964 Monroe Wall and associates, working at the Research Triangle Institute under a National Cancer Institute (NCI) contract, isolated the active compound in paclitaxel from the bark of the common yew tree [2]. Its tortuous development, complicated by difficulties in material procurement, compound purification and formulation, delayed its entry into clinical trials until 1983, and its efficacy in treating ovarian cancer was not demonstrated until 1987 [3].
    [Show full text]
  • Incentivizing the Utilization of Pharmacogenomics in Drug Development Valerie Gutmann Koch
    Journal of Health Care Law and Policy Volume 15 | Issue 2 Article 3 Incentivizing the Utilization of Pharmacogenomics in Drug Development Valerie Gutmann Koch Follow this and additional works at: http://digitalcommons.law.umaryland.edu/jhclp Part of the Chemicals and Drugs Commons, and the Health Law Commons Recommended Citation Valerie G. Koch, Incentivizing the Utilization of Pharmacogenomics in Drug Development, 15 J. Health Care L. & Pol'y 263 (2012). Available at: http://digitalcommons.law.umaryland.edu/jhclp/vol15/iss2/3 This Article is brought to you for free and open access by DigitalCommons@UM Carey Law. It has been accepted for inclusion in Journal of Health Care Law and Policy by an authorized administrator of DigitalCommons@UM Carey Law. For more information, please contact [email protected]. INCENTIVIZING THE UTILIZATION OF PHARMACOGENOMICS IN DRUG DEVELOPMENT VALERIE GUTMANN KOCH* I. INTRODUCTION The last decades have witnessed remarkable advancements in the fields of genetics and genomics, highlighted by the successful completion of the map of the human genome in 2003.' With this achievement came scientific possibilities that, only a few decades earlier, seemed more science fiction than reality. Of these developments, pharmacogenomics is hailed by many as a panacea for problems associated with pharmaceutical drug use and development.2 The Human Genome Project (HGP) and associated research have demonstrated that all human beings share 99.9 percent of their DNA. 3 Pharmacogenomics focuses on the 0.1 percent differences between individuals and promises to allow physicians to tailor a patient's prescription according to his or her genetic profile, reducing painful and sometimes deadly side effects, ensuring appropriate dosage decisions, and targeting specific disease pathways.4 Copyright © 2012 by Valerie Gutmann Koch.
    [Show full text]
  • Computer Aided Drug Design: a Novel Loom to Drug Discovery
    Organic and Medicinal Chemistry International Journal ISSN 2474-7610 Review Article Organic & Medicinal Chem IJ Volume 1 Issue 4 - February 2017 Copyright © All rights are reserved by Syed Sarim Imam DOI: 10.19080/OMCIJ.2017.01.555567 Computer Aided Drug Design: A Novel Loom To Drug Discovery Syed Sarim Imam and Sadaf Jamal Gilani* Department of Pharmaceutics, Glocal School of Pharmacy, The Glocal University, Saharanpur, U.P, India Department of Pharmaceutical Chemistry, Glocal School of Pharmacy, The Glocal University, Saharanpur, U.P, India Submission: February 15, 2017; Published: February 20, 2017 *Corresponding author: Dr. Sadaf Jamal Gilani, Associate Professor, Department of Pharmaceutical Chemistry, Glocal School of Pharmacy, The Glocal University, Saharanpur-247121, Uttar Pradesh, India, Tel: ; Email: Abstract Computer-Aided Drug Design (CADD) is a growing effort to apply computational power to the combined chemical and biological space in order to streamline drug discovery design, development, and optimization. This technique of drug discovery and development are very time and resources consuming processes. But this tool can act as a virtual shortcut, assisting in the expedition of this long process and potentially reducing the cost of research and development. This application has been widely used in the biomedical arena, in silico design is being utilized Here in this mini review, we present overviews of computational methods used in the different arena of drug discovery and focusing some of theto expedite recent successes. and facilitate hit identification, hit-to-lead selection, optimize the pharmacokinetic profile, toxicity profile and avoid safety issues. Keywords: Computer-aided drug design (CADD); In-silico design; Computational methods Introduction b.
    [Show full text]
  • Pharmacogenomic-Guided Drug Development LJ Lesko and J Woodcock 21
    The Pharmacogenomics Journal (2002) 2, 20–24 2002 Nature Publishing Group All rights reserved 1470-269X/02 $25.00 www.nature.com/tpj EELS (Ethical, Economic, Legal & Social) ARTICLE response, caused by genetic differ- Pharmacogenomic-guided drug ences. The ability to predict and account for such differences could development: regulatory perspective markedly improve the therapeutic index of many drug interventions. LJ Lesko1 and J Woodcock2 Finally, it is hoped that genetically- based mechanisms of toxicity can be 1Office of Clinical Pharmacology and Biopharmaceutics, Center for Drug Evaluation and elucidated, and adverse effects avoided, Research, Food and Drug Administration, Rockville, MD, USA; 2Office of the Center by application of pharmacogenomic Director, Center for Drug Evaluation and Research, Food and Drug Administration, information. Rockville, MD, USA The explosion of interest in PGt and PGx has raised concerns that the regu- latory environment could inhibit pro- The Pharmacogenomics Journal (2002) 2, mechanism of drug actions, and the gress. While drug discovery and pre- 20–24. DOI: 10.1038/sj/tpj/ 6500046 drug–disease interaction. Taken clinical studies are not likely to be together, these techniques are significantly affected, there is concern Pharmacogenetics (PGt) and now the expected to yield major advances in about the use of PGt/PGx in clinical more global term, pharmacogenomics identifying drug candidates. trials. This article provides an overall (PGx), have come to the forefront after Genomic information will be regulatory perspective on the clinical an evolutionary period of more than increasingly used in the preclinical study issues and considerations, many 30 years. Several transforming events phases of drug development.
    [Show full text]
  • Topic 7 Drug Discovery and Design Pt. 1
    Topic 7 Drug Discovery and design Pt. 1 Chapter 9 Patrick Contents Part 1: Sections 9.1-9.3 1. Target disease 2. Drug Targets 3. Testing Drugs 3.1. In vivo Tests 3.2. In vitro Tests 3.2.1. Enzyme Inhibition Tests 3.2.2. Testing with Receptors DRUG DESIGN AND DEVELOPMENT Stages 1) Identify target disease 2) Identify drug target 3) Establish testing procedures 4) Find a lead compound 5) Structure Activity Relationships (SAR) 6) Identify a pharmacophore 7) Drug design- optimising target interactions 8) Drug design - optimising pharmacokinetic properties 9) Toxicological and safety tests 10) Chemical development and production 11) Patenting and regulatory affairs 12) Clinical trials 1. TARGET DISEASE Priority for the Pharmaceutical Industry • Can the profits from marketing a new drug outweigh the cost of developing and testing that drug? Questions to be addressed • Is the disease widespread? (e.g. cardiovascular disease, ulcers, malaria) • Does the disease affect the first world? (e.g. cardiovascular disease, ulcers) • Are there drugs already on the market? • If so, what are there advantages and disadvantages? (e.g. side effects) • Can one identify a market advantage for a new therapy? 2. DRUG TARGETS-Remember? A) LIPIDS Cell Membrane Lipids B) PROTEINS Receptors Enzymes Carrier Proteins Structural Proteins (tubulin) C) NUCLEIC ACIDS DNA RNA D) CARBOHYDRATES Cell surface carbohydrates Antigens and recognition molecules 2. DRUG TARGETS TARGET SELECTIVITY Between species • Antibacterial and antiviral agents • Identify targets which are unique to the invading pathogen • Identify targets which are shared but which are significantly different in structure Within the body • Selectivity between different enzymes, receptors etc.
    [Show full text]
  • Hit Discovery and Hit-To-Lead Approaches Reviews
    Drug Discovery Today Volume 11, Numbers 15/16 August 2006 REVIEWS POST SCREEN Hit discovery and hit-to-lead approaches Reviews Gyo¨ rgy M. Keseru˝ 1 and Gergely M. Makara2 1 CADD&HTS Unit, Gedeon Richter Ltd, 19-21 Gyo¨mro˝iu´t, Budapest, H-1103, Hungary 2 Merck Research Laboratories, Merck & Co, RY80Y-325, 126 E. Lincoln Ave, Rahway, New Jersey, 07065, USA Hit discovery technologies range from traditional high-throughput screening to affinity selection of large libraries, fragment-based techniques and computer-aided de novo design, many of which have been extensively reviewed. Development of quality leads using hit confirmation and hit-to-lead approaches present their own challenges, depending on the hit discovery method used to identify the initial hits. In this paper, we summarize common industry practices adopted to tackle hit-to-lead challenges and review how the advantages and drawbacks of different hit discovery techniques could affect the various issues hit-to-lead groups face. It has been shown that marketed drugs are very frequently highly All hit discovery approaches have – often orthogonal – short- similar to the leads from which they were derived [1]. Thus, both comings prompting the frequent use of multiple techniques for hit the quality and the quantity of lead classes available to medicinal confirmation (Table 1). Challenges facing traditional HTS tech- chemists are primary drivers for discovering best-in-class medi- nologies include high false-positive rates, the need for reporter cines. This makes lead generation a crucial step in the drug dis- assays and the limitation in throughput imposed by testing com- covery process.
    [Show full text]