1
|
Freuville L, Matthys C, Quinton L, Gillet JP. Venom-derived peptides for breaking through the glass ceiling of drug development. Front Chem 2024; 12:1465459. [PMID: 39398192 PMCID: PMC11468230 DOI: 10.3389/fchem.2024.1465459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024] Open
Abstract
Venoms are complex mixtures produced by animals and consist of hundreds of components including small molecules, peptides, and enzymes selected for effectiveness and efficacy over millions of years of evolution. With the development of venomics, which combines genomics, transcriptomics, and proteomics to study animal venoms and their effects deeply, researchers have identified molecules that selectively and effectively act against membrane targets, such as ion channels and G protein-coupled receptors. Due to their remarkable physico-chemical properties, these molecules represent a credible source of new lead compounds. Today, not less than 11 approved venom-derived drugs are on the market. In this review, we aimed to highlight the advances in the use of venom peptides in the treatment of diseases such as neurological disorders, cardiovascular diseases, or cancer. We report on the origin and activity of the peptides already approved and provide a comprehensive overview of those still in development.
Collapse
Affiliation(s)
- Lou Freuville
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Chloé Matthys
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| | - Loïc Quinton
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Jean-Pierre Gillet
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| |
Collapse
|
2
|
Oganesyan I, Jenkins TP, Laustsen AH, Zenobi R, Harrison JA. Streamlining the Analysis of Proteins from Snake Venom. J Proteome Res 2024; 23:2367-2375. [PMID: 38814071 DOI: 10.1021/acs.jproteome.4c00013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Investigating snake venom is necessary for developing new treatments for envenoming and harnessing the therapeutic potential that lies within venom toxins. Despite considerable efforts in previous research, several technical challenges remain for characterizing the individual components within such complex mixtures. Here, we present native and top-down mass spectrometry (MS) workflows that enable the analysis of individual venom proteins within complex mixtures and showcase the utility of these methodologies on King cobra (Ophiophagus hannah) venom. First, we coupled ion mobility spectrometry for separation and electron capture dissociation for charge reduction to resolve highly convoluted mass spectra containing multiple proteins with masses ranging from 55 to 127 kDa. Next, we performed a top-down glycomic analysis of a 25.5 kDa toxin, showing that this protein contains a fucosylated complex glycan. Finally, temperature-controlled nanoelectrospray mass spectrometry facilitated the top-down sequence analysis of a β-cardiotoxin, which cannot be fragmented by collisional energy due to its disulfide bond pattern. The work presented here demonstrates the applicability of new and promising MS methods for snake venom analysis.
Collapse
Affiliation(s)
- Irina Oganesyan
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Timothy P Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
- VenomAid Diagnostics ApS, DK-2800 Kongens Lyngby, Denmark
| | - Renato Zenobi
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Julian A Harrison
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland
| |
Collapse
|
3
|
Offor BC, Piater LA. Snake venom toxins: Potential anticancer therapeutics. J Appl Toxicol 2024; 44:666-685. [PMID: 37697914 DOI: 10.1002/jat.4544] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/21/2023] [Accepted: 08/29/2023] [Indexed: 09/13/2023]
Abstract
Snake venom contains a cocktail of compounds dominated by proteins and peptides, which make up the toxin. The toxin components of snake venom attack several targets in the human body including the neuromuscular system, kidney and blood coagulation system and cause pathologies. As such, the venom toxins can be managed and used for the treatment of these diseases. In this regard, Captopril used in the treatment of cardiovascular diseases was the first animal venom toxin-based drug approved by the US Food and Drug Administration and the European Medicines Agency. Cancers cause morbidity and mortality worldwide. Due to side effects associated with the current cancer treatments including chemotherapy, radiotherapy, immunotherapy, hormonal therapy and surgery, there is a need to improve the efficacy of current treatments and/or develop novel drugs from natural sources including animal toxin-based drugs. There is a long history of earlier and ongoing studies implicating snake venom toxins as potential anticancer therapies. Here, we review the role of crude snake venoms and toxins including phospholipase A2, L-amino acid oxidase, C-type lectin and disintegrin as potential anticancer agents tested in cancer cell lines and animal tumour models in comparison to normal cell lines. Some of the anti-tumour activities of snake venom toxins include induction of cytotoxicity, apoptosis, cell cycle arrest and inhibition of metastasis, angiogenesis and tumour growth. We thus propose the advancement of multidisciplinary approaches to more pre-clinical and clinical studies for enhanced bioavailability and targeted delivery of snake venom toxin-based anticancer drugs.
Collapse
Affiliation(s)
- Benedict C Offor
- Department of Biochemistry, University of Johannesburg, Auckland Park, South Africa
| | - Lizelle A Piater
- Department of Biochemistry, University of Johannesburg, Auckland Park, South Africa
| |
Collapse
|
4
|
Nargund R, Wyvratt M, Lin S, Sebhat I, Greenlee W. Annotated Bibliography of Dr. Arthur A. Patchett. J Med Chem 2023; 66:15567-15575. [PMID: 38032081 DOI: 10.1021/acs.jmedchem.3c02131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
|
5
|
Almeida GDO, de Oliveira IS, Arantes EC, Sampaio SV. Snake venom disintegrins update: insights about new findings. J Venom Anim Toxins Incl Trop Dis 2023; 29:e20230039. [PMID: 37818211 PMCID: PMC10561651 DOI: 10.1590/1678-9199-jvatitd-2023-0039] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/25/2023] [Indexed: 10/12/2023] Open
Abstract
Snake venom disintegrins are low molecular weight, non-enzymatic proteins rich in cysteine, present in the venom of snakes from the families Viperidae, Crotalidae, Atractaspididae, Elapidae, and Colubridae. This family of proteins originated in venom through the proteolytic processing of metalloproteinases (SVMPs), which, in turn, evolved from a gene encoding an A Disintegrin And Metalloprotease (ADAM) molecule. Disintegrins have a recognition motif for integrins in their structure, allowing interaction with these transmembrane adhesion receptors and preventing their binding to proteins in the extracellular matrix and other cells. This interaction gives disintegrins their wide range of biological functions, including inhibition of platelet aggregation and antitumor activity. As a result, many studies have been conducted in an attempt to use these natural compounds as a basis for developing therapies for the treatment of various diseases. Furthermore, the FDA has approved Tirofiban and Eptifibatide as antiplatelet compounds, and they are synthesized from the structure of echistatin and barbourin, respectively. In this review, we discuss some of the main functional and structural characteristics of this class of proteins and their potential for therapeutic use.
Collapse
Affiliation(s)
- Gabriela de Oliveira Almeida
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Isadora Sousa de Oliveira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Eliane Candiani Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Suely Vilela Sampaio
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
6
|
Abdullaev MG. New Palladium-Containing Polymer Catalysts for the Synthesis of Enalapril. Pharm Chem J 2023. [DOI: 10.1007/s11094-023-02801-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
7
|
Ageitos L, Torres MDT, de la Fuente-Nunez C. Biologically Active Peptides from Venoms: Applications in Antibiotic Resistance, Cancer, and Beyond. Int J Mol Sci 2022; 23:ijms232315437. [PMID: 36499761 PMCID: PMC9740984 DOI: 10.3390/ijms232315437] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 12/12/2022] Open
Abstract
Peptides are potential therapeutic alternatives against global diseases, such as antimicrobial-resistant infections and cancer. Venoms are a rich source of bioactive peptides that have evolved over time to act on specific targets of the prey. Peptides are one of the main components responsible for the biological activity and toxicity of venoms. South American organisms such as scorpions, snakes, and spiders are important producers of a myriad of peptides with different biological activities. In this review, we report the main venom-derived peptide families produced from South American organisms and their corresponding activities and biological targets.
Collapse
Affiliation(s)
- Lucía Ageitos
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcelo D. T. Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence:
| |
Collapse
|
8
|
Calcatelli A, Denton RM, Ball LT. Modular Synthesis of α,α-Diaryl α-Amino Esters via Bi(V)-Mediated Arylation/S N2-Displacement of Kukhtin–Ramirez Intermediates. Org Lett 2022; 24:8002-8007. [PMID: 36278869 PMCID: PMC9641671 DOI: 10.1021/acs.orglett.2c03201] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
We report a concise and modular approach to α,α-diaryl
α-amino esters from readily available α-keto esters. This
mild, one-pot protocol proceeds via ketone umpolung, with in situ formation of a Kukhtin–Ramirez intermediate
preceding sequential electrophilic arylation by Bi(V) and SN2 displacement by an amine. The methodology is compatible with a
wide range of anilines and primary amines - including derivatives
of drugs and proteinogenic amino acids - Bi(V) arylating agents, and
α-keto ester substrates.
Collapse
Affiliation(s)
| | - Ross M. Denton
- School of Chemistry, University of Nottingham, Nottingham NG7 2RD, U.K
- GlaxoSmithKline Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham, 6 Triumph Road, Nottingham NG7 2GA, U.K
| | - Liam T. Ball
- School of Chemistry, University of Nottingham, Nottingham NG7 2RD, U.K
| |
Collapse
|
9
|
Oliveira AL, Viegas MF, da Silva SL, Soares AM, Ramos MJ, Fernandes PA. The chemistry of snake venom and its medicinal potential. Nat Rev Chem 2022; 6:451-469. [PMID: 35702592 PMCID: PMC9185726 DOI: 10.1038/s41570-022-00393-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 12/15/2022]
Abstract
The fascination and fear of snakes dates back to time immemorial, with the first scientific treatise on snakebite envenoming, the Brooklyn Medical Papyrus, dating from ancient Egypt. Owing to their lethality, snakes have often been associated with images of perfidy, treachery and death. However, snakes did not always have such negative connotations. The curative capacity of venom has been known since antiquity, also making the snake a symbol of pharmacy and medicine. Today, there is renewed interest in pursuing snake-venom-based therapies. This Review focuses on the chemistry of snake venom and the potential for venom to be exploited for medicinal purposes in the development of drugs. The mixture of toxins that constitute snake venom is examined, focusing on the molecular structure, chemical reactivity and target recognition of the most bioactive toxins, from which bioactive drugs might be developed. The design and working mechanisms of snake-venom-derived drugs are illustrated, and the strategies by which toxins are transformed into therapeutics are analysed. Finally, the challenges in realizing the immense curative potential of snake venom are discussed, and chemical strategies by which a plethora of new drugs could be derived from snake venom are proposed.
Collapse
Affiliation(s)
- Ana L. Oliveira
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Matilde F. Viegas
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Saulo L. da Silva
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Andreimar M. Soares
- Biotechnology Laboratory for Proteins and Bioactive Compounds from the Western Amazon, Oswaldo Cruz Foundation, National Institute of Epidemiology in the Western Amazon (INCT-EpiAmO), Porto Velho, Brazil
- Sao Lucas Universitary Center (UniSL), Porto Velho, Brazil
| | - Maria J. Ramos
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Pedro A. Fernandes
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| |
Collapse
|
10
|
Smallwood TB, Clark RJ. Advances in venom peptide drug discovery: where are we at and where are we heading? Expert Opin Drug Discov 2021; 16:1163-1173. [PMID: 33914674 DOI: 10.1080/17460441.2021.1922386] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: Animal venoms are a complex mixture of bioactive molecules that have evolved over millions of years for prey capture and defense from predators. Venom consists of many different types of molecules, with disulfide-rich peptides being a major component in most venoms. The study of these potent and highly selective molecules has led to the development of venom-derived drugs for diseases such as type 2 diabetes mellitus and chronic pain. As technologies have improved, more bioactive peptides have been discovered from venomous animals. Many of these molecules may have applications as tools for understanding normal and disease physiology, therapeutics, cosmetics or in agriculture.Areas covered: This article reviews venom-derived drugs approved by the FDA and venom-derived peptides currently in development. It discusses the challenges faced by venom-derived peptide drugs during drug development and the future for venom-derived peptides.Expert opinion: New techniques such as toxin driven discovery are expanding the pipeline of venom-derived peptides. There are many venom-derived peptides currently in preclinical and clinical trials that would have remained undiscovered using traditional approaches. A renewed focus on venoms, with advances in technology, will broaden the diversity of venom-derived peptide therapeutics and expand our knowledge of their molecular targets.
Collapse
Affiliation(s)
- Taylor B Smallwood
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Richard J Clark
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| |
Collapse
|
11
|
Albat D, Neudörfl J, Schmalz H. A General Stereocontrolled Synthesis of Opines through Asymmetric Pd‐Catalyzed N‐Allylation of Amino Acid Esters. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Dominik Albat
- Department of Chemistry University of Cologne Greinstrasse 4 50939 Köln Germany
| | | | | |
Collapse
|
12
|
Coulter-Parkhill A, McClean S, Gault VA, Irwin N. Therapeutic Potential of Peptides Derived from Animal Venoms: Current Views and Emerging Drugs for Diabetes. Clin Med Insights Endocrinol Diabetes 2021; 14:11795514211006071. [PMID: 34621137 PMCID: PMC8491154 DOI: 10.1177/11795514211006071] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/10/2021] [Indexed: 12/13/2022] Open
Abstract
The therapeutic potential of venom-derived drugs is evident today. Currently, several significant drugs are FDA approved for human use that descend directly from animal venom products, with others having undergone, or progressing through, clinical trials. In addition, there is growing awareness of the important cosmeceutical application of venom-derived products. The success of venom-derived compounds is linked to their increased bioactivity, specificity and stability when compared to synthetically engineered compounds. This review highlights advancements in venom-derived compounds for the treatment of diabetes and related disorders. Exendin-4, originating from the saliva of Gila monster lizard, represents proof-of-concept for this drug discovery pathway in diabetes. More recent evidence emphasises the potential of venom-derived compounds from bees, cone snails, sea anemones, scorpions, snakes and spiders to effectively manage glycaemic control. Such compounds could represent exciting exploitable scaffolds for future drug discovery in diabetes, as well as providing tools to allow for a better understanding of cell signalling pathways linked to insulin secretion and metabolism.
Collapse
Affiliation(s)
| | | | - Victor A Gault
- Diabetes Research Group, Ulster University, Coleraine, UK
| | - Nigel Irwin
- Diabetes Research Group, Ulster University, Coleraine, UK
| |
Collapse
|
13
|
Hydrogenation N-alkylation of α-alanyl-α-proline with ethyl 2-oxo-4-phenylbutanoate on organometallic catalysts. Russ Chem Bull 2020. [DOI: 10.1007/s11172-020-2979-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
14
|
Bordon KDCF, Cologna CT, Fornari-Baldo EC, Pinheiro-Júnior EL, Cerni FA, Amorim FG, Anjolette FAP, Cordeiro FA, Wiezel GA, Cardoso IA, Ferreira IG, de Oliveira IS, Boldrini-França J, Pucca MB, Baldo MA, Arantes EC. From Animal Poisons and Venoms to Medicines: Achievements, Challenges and Perspectives in Drug Discovery. Front Pharmacol 2020; 11:1132. [PMID: 32848750 PMCID: PMC7396678 DOI: 10.3389/fphar.2020.01132] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022] Open
Abstract
Animal poisons and venoms are comprised of different classes of molecules displaying wide-ranging pharmacological activities. This review aims to provide an in-depth view of toxin-based compounds from terrestrial and marine organisms used as diagnostic tools, experimental molecules to validate postulated therapeutic targets, drug libraries, prototypes for the design of drugs, cosmeceuticals, and therapeutic agents. However, making these molecules applicable requires extensive preclinical trials, with some applications also demanding clinical trials, in order to validate their molecular target, mechanism of action, effective dose, potential adverse effects, as well as other fundamental parameters. Here we go through the pitfalls for a toxin-based potential therapeutic drug to become eligible for clinical trials and marketing. The manuscript also presents an overview of the current picture for several molecules from different animal venoms and poisons (such as those from amphibians, cone snails, hymenopterans, scorpions, sea anemones, snakes, spiders, tetraodontiformes, bats, and shrews) that have been used in clinical trials. Advances and perspectives on the therapeutic potential of molecules from other underexploited animals, such as caterpillars and ticks, are also reported. The challenges faced during the lengthy and costly preclinical and clinical studies and how to overcome these hindrances are also discussed for that drug candidates going to the bedside. It covers most of the drugs developed using toxins, the molecules that have failed and those that are currently in clinical trials. The article presents a detailed overview of toxins that have been used as therapeutic agents, including their discovery, formulation, dosage, indications, main adverse effects, and pregnancy and breastfeeding prescription warnings. Toxins in diagnosis, as well as cosmeceuticals and atypical therapies (bee venom and leech therapies) are also reported. The level of cumulative and detailed information provided in this review may help pharmacists, physicians, biotechnologists, pharmacologists, and scientists interested in toxinology, drug discovery, and development of toxin-based products.
Collapse
Affiliation(s)
- Karla de Castro Figueiredo Bordon
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Camila Takeno Cologna
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Ernesto Lopes Pinheiro-Júnior
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe Augusto Cerni
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernanda Gobbi Amorim
- Postgraduate Program in Pharmaceutical Sciences, Vila Velha University, Vila Velha, Brazil
| | | | - Francielle Almeida Cordeiro
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Gisele Adriano Wiezel
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Iara Aimê Cardoso
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isabela Gobbo Ferreira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isadora Sousa de Oliveira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | - Mateus Amaral Baldo
- Health and Science Institute, Paulista University, São José do Rio Pardo, Brazil
| | - Eliane Candiani Arantes
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
15
|
Kumar S, Jaipuri FA, Waldo JP, Potturi H, Marcinowicz A, Adams J, Van Allen C, Zhuang H, Vahanian N, Link C, Brincks EL, Mautino MR. Discovery of indoximod prodrugs and characterization of clinical candidate NLG802. Eur J Med Chem 2020; 198:112373. [PMID: 32422549 DOI: 10.1016/j.ejmech.2020.112373] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/20/2022]
Abstract
A series of different prodrugs of indoximod, including estesrs and peptide amides were synthesized with the aim of improving its oral bioavailability in humans. The pharmacokinetics of prodrugs that were stable in buffers, plasma and simulated gastric and intestinal fluids was first assessed in rats after oral dosing in solution or in capsule formulation. Two prodrugs that produced the highest exposure to indoximod in rats were further tested in Cynomolgus monkeys, a species in which indoximod has oral bioavailability of 6-10% and an equivalent dose-dependent exposure profile as humans. NLG802 was selected as the clinical development candidate after increasing oral bioavailability (>5-fold), Cmax (6.1-3.6 fold) and AUC (2.9-5.2 fold) in monkeys, compared to equivalent molar oral doses of indoximod. NLG802 is extensively absorbed and rapidly metabolized to indoximod in all species tested and shows a safe toxicological profile at the anticipated therapeutic doses. NLG802 markedly enhanced the anti-tumor responses of tumor-specific pmel-1 T cells in a melanoma tumor model. In conclusion, NLG802 is a prodrug of indoximod expected to increase clinical drug exposure to indoximod above the current achievable levels, thus increasing the possibility of therapeutic effects in a larger fraction of the target patient population.
Collapse
Affiliation(s)
| | | | | | - Hima Potturi
- NewLink Genetics, Ames, IA, 50010, United States
| | | | - James Adams
- NewLink Genetics, Ames, IA, 50010, United States
| | | | - Hong Zhuang
- NewLink Genetics, Ames, IA, 50010, United States
| | | | - Charles Link
- NewLink Genetics, Ames, IA, 50010, United States
| | - Erik L Brincks
- NewLink Genetics, Ames, IA, 50010, United States; Lumos Pharma, Inc., Ames, IA, 50010, United States.
| | | |
Collapse
|
16
|
Verbeeck RK, Kanfer I, Löbenberg R, Abrahamsson B, Cristofoletti R, Groot DW, Langguth P, Polli JE, Parr A, Shah VP, Mehta M, Dressman JB. Biowaiver Monographs for Immediate-Release Solid Oral Dosage Forms: Enalapril. J Pharm Sci 2017; 106:1933-1943. [PMID: 28435140 DOI: 10.1016/j.xphs.2017.04.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 04/10/2017] [Accepted: 04/12/2017] [Indexed: 11/30/2022]
Abstract
Literature data relevant to the decision to allow a waiver of in vivo bioequivalence testing for the marketing authorization of immediate-release, solid oral dosage forms containing enalapril maleate are reviewed. Enalapril, a prodrug, is hydrolyzed by carboxylesterases to the active angiotensin-converting enzyme inhibitor enalaprilat. Enalapril as the maleate salt is shown to be highly soluble, but only 60%-70% of an orally administered dose of enalapril is absorbed from the gastrointestinal tract into the enterocytes. Consequently, enalapril maleate is a Biopharmaceutics Classification System class III substance. Because in situ conversion of the maleate salt to the sodium salt is sometimes used in production of the finished drug product, not every enalapril maleate-labeled finished product actually contains the maleate salt. Enalapril is not considered to have a narrow therapeutic index. With this background, a biowaiver-based approval procedure for new generic products or after major revisions to existing products is deemed acceptable, provided the in vitro dissolution of both test and reference preparation is very rapid (at least 85% within 15 min at pH 1.2, 4.5, and 6.8). Additionally, the test and reference product must contain the identical active drug ingredient.
Collapse
Affiliation(s)
- Roger K Verbeeck
- School of Pharmacy, University of Namibia, Windhoek, Namibia; Faculty of Pharmacy, Rhodes University, Grahamstown, South Africa
| | - Isadore Kanfer
- Faculty of Pharmacy, Rhodes University, Grahamstown, South Africa; Lesley Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Raimar Löbenberg
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | | | - Rodrigo Cristofoletti
- Division of Bioequivalence, Brazilian Health Surveillance Agency (ANVISA), Brasılia, Brazil
| | - D W Groot
- RIVM-National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Peter Langguth
- Institut für Pharmazie und Biochemie, Johannes Gutenberg-Universität Mainz, Mainz, Germany
| | - James E Polli
- School of Pharmacy, University of Maryland, Baltimore, Maryland 21201
| | - Alan Parr
- BioCeutics LLC, Raleigh-Durham, North Carolina 27501
| | - Vinod P Shah
- International Pharmaceutical Federation FIP, The Hague, The Netherlands
| | - Mehul Mehta
- United States Food and Drug Administration, Centre for Drug Evaluation and Research, Silver Spring, Maryland 20993
| | - Jennifer B Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany.
| |
Collapse
|
17
|
Antza C, Stabouli S, Kotsis V. Combination therapy with lercanidipine and enalapril in the management of the hypertensive patient: an update of the evidence. Vasc Health Risk Manag 2016; 12:443-451. [PMID: 27895487 PMCID: PMC5118038 DOI: 10.2147/vhrm.s91020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hypertension is an important risk factor for premature death as it increases the probability of stroke, myocardial infarction, and heart failure. Antihypertensive drugs can decrease cardiovascular (CV) morbidity and mortality. The majority of hypertensive patients need more than one antihypertensive agent to attain blood pressure (BP) targets. Monotherapy can effectively reduce BP only in 20%–40% of patients. Multiple mechanisms including increased peripheral vascular resistance, increased cardiac work, and hypervolemia are involved in the pathogenesis of hypertension. Targeting multiple pathways may more potently reduce BP. Increasing the dose of a single agent in many cases does not provide the expected BP-lowering effect because the underlying mechanism of the BP increase is either different or already corrected with the lower dose. Moreover, drugs acting on different pathways may have synergistic effects and thus better control hypertension. It is well known that diuretics enhance the actions of renin–angiotensin aldosterone system and activate it as a feedback to the reduced circulated blood volume. The addition of a renin–angiotensin aldosterone system blocker to a diuretic may more effectively reduce BP because the system is upregulated. Reducing the maximal dose of an agent may also reduce possible side effects if they are dose dependent. The increased prevalence of peripheral edema with higher doses of calcium channel blockers (CCBs) is reduced when renin–angiotensin aldosterone system blockers are added to CCBs through vein dilation. The effectiveness of the combination of enalapril with lercanidipine in reducing BP, the safety profile, and the use of the combination of angiotensin-converting enzyme inhibitors with CCBs in clinical trials with excellent CV hard end point outcomes make this combination a promising therapy in the treatment of hypertension.
Collapse
Affiliation(s)
- Christina Antza
- Hypertension Center, Third Department of Medicine, Papageorgiou Hospital
| | - Stella Stabouli
- First Department of Pediatrics, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vasilios Kotsis
- Hypertension Center, Third Department of Medicine, Papageorgiou Hospital
| |
Collapse
|
18
|
Sulfur-Containing Angiotensin-Converting Enzyme Inhibitor 3-Thienylalanine-Ornithyl-Proline Activates Endothelial Function and Expression of Genes Involved in Renin–Angiotensin System. J Cardiovasc Pharmacol 2013; 61:311-7. [DOI: 10.1097/fjc.0b013e318280e16e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
Won CS, Oberlies NH, Paine MF. Mechanisms underlying food-drug interactions: inhibition of intestinal metabolism and transport. Pharmacol Ther 2012; 136:186-201. [PMID: 22884524 DOI: 10.1016/j.pharmthera.2012.08.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 07/23/2012] [Indexed: 12/21/2022]
Abstract
Food-drug interaction studies are critical to evaluate appropriate dosing, timing, and formulation of new drug candidates. These interactions often reflect prandial-associated changes in the extent and/or rate of systemic drug exposure. Physiologic and physicochemical mechanisms underlying food effects on drug disposition are well-characterized. However, biochemical mechanisms involving drug metabolizing enzymes and transport proteins remain underexplored. Several plant-derived beverages have been shown to modulate enzymes and transporters in the intestine, leading to altered pharmacokinetic (PK) and potentially negative pharmacodynamic (PD) outcomes. Commonly consumed fruit juices, teas, and alcoholic drinks contain phytochemicals that inhibit intestinal cytochrome P450 and phase II conjugation enzymes, as well as uptake and efflux transport proteins. Whereas myriad phytochemicals have been shown to inhibit these processes in vitro, translation to the clinic has been deemed insignificant or undetermined. An overlooked prerequisite for elucidating food effects on drug PK is thorough knowledge of causative bioactive ingredients. Substantial variability in bioactive ingredient composition and activity of a given dietary substance poses a challenge in conducting robust food-drug interaction studies. This confounding factor can be addressed by identifying and characterizing specific components, which could be used as marker compounds to improve clinical trial design and quantitatively predict food effects. Interpretation and integration of data from in vitro, in vivo, and in silico studies require collaborative expertise from multiple disciplines, from botany to clinical pharmacology (i.e., plant to patient). Development of more systematic methods and guidelines is needed to address the general lack of information on examining drug-dietary substance interactions prospectively.
Collapse
Affiliation(s)
- Christina S Won
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7569, USA
| | | | | |
Collapse
|
20
|
Koh CY, Kini RM. From snake venom toxins to therapeutics – Cardiovascular examples. Toxicon 2012; 59:497-506. [DOI: 10.1016/j.toxicon.2011.03.017] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 03/15/2011] [Accepted: 03/16/2011] [Indexed: 11/30/2022]
|
21
|
Won CS, Oberlies NH, Paine MF. Influence of dietary substances on intestinal drug metabolism and transport. Curr Drug Metab 2011; 11:778-92. [PMID: 21189136 DOI: 10.2174/138920010794328869] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 11/20/2010] [Indexed: 11/22/2022]
Abstract
Successful delivery of promising new chemical entities via the oral route is rife with challenges, some of which cannot be explained or foreseen during drug development. Further complicating an already multifaceted problem is the obvious, yet often overlooked, effect of dietary substances on drug disposition and response. Some dietary substances, particularly fruit juices, have been shown to inhibit biochemical processes in the intestine, leading to altered pharmacokinetic (PK), and potentially pharmacodynamic (PD), outcomes. Inhibition of intestinal CYP3Amediated metabolism is the major mechanism by which fruit juices, including grapefruit juice, enhances systemic exposure to new and already marketed drugs. Inhibition of intestinal non-CYP3A enzymes and apically-located transport proteins represent recently identified mechanisms that can alter PK and PD. Several fruit juices have been shown to inhibit these processes in vitro, but some interactions have not translated to the clinic. The lack of in vitroin vivo concordance is due largely to a lack of rigorous methods to elucidate causative ingredients prior to clinical testing. Identification of specific components and underlying mechanisms is challenging, as dietary substances frequently contain multiple, often unknown, bioactive ingredients that vary in composition and bioactivity. A translational research approach, combining expertise from clinical pharmacologists and natural products chemists, is needed to develop robust models describing PK/PD relationships between a given dietary substance and drug of interest. Validation of these models through well-designed clinical trials would facilitate development of common practice guidelines for managing drug-dietary substance interactions appropriately.
Collapse
Affiliation(s)
- Christina S Won
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7569, USA
| | | | | |
Collapse
|
22
|
Abstract
Numerous metalloproteins are important therapeutic targets that are gaining increased attention in the medicinal and bioinorganic chemistry communities. This Perspective article describes some emerging trends and recent findings in the area of metalloprotein inhibitor discovery and development. In particular, increasing recognition of the importance of the metal-ligand interactions in these systems calls for more input and consideration from the bioinorganic community to address questions traditionally confined to the medicinal chemistry community.
Collapse
Affiliation(s)
| | - Seth M. Cohen
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093-0358, U.S.A. Fax: 858-822-5598; Tel: 858-822-5596
| |
Collapse
|
23
|
Toporišič R, Mlakar A, Hvala J, Prislan I, Zupančič-Kralj L. Identification of new impurities of enalapril maleate on oxidation in the presence of magnesium monoperoxyphthalate. J Pharm Biomed Anal 2010; 52:294-9. [DOI: 10.1016/j.jpba.2009.12.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 12/06/2009] [Accepted: 12/18/2009] [Indexed: 11/27/2022]
|
24
|
Effect of 3-Thienylalanine-Ornithine-Proline, New Sulfur-containing Angiotensin-converting Enzyme Inhibitor on Blood Pressure and Oxidative Stress in Spontaneously Hypertensive Rats. J Cardiovasc Pharmacol 2009; 53:145-50. [DOI: 10.1097/fjc.0b013e318197c616] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Abstract
A recent study challenges the oft-held notion that ester bonds in prodrug molecules are cleaved rapidly and completely inside cells by endogenous, nonspecific esterases. Structure-activity relationship studies on acylated sugars reveal that regioisomeric compounds display disparate biological activity, suggesting that ester bonds can persist in a cellular context.
Collapse
Affiliation(s)
- Luke D. Lavis
- Janelia Farm Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, Virginia 20147
| |
Collapse
|
26
|
Dimitropoulou C, Chatterjee A, McCloud L, Yetik-Anacak G, Catravas JD. Angiotensin, bradykinin and the endothelium. Handb Exp Pharmacol 2007:255-94. [PMID: 16999222 DOI: 10.1007/3-540-32967-6_8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Angiotensins and kinins are endogenous peptides with diverse biological actions; as such, they represent current and future targets of therapeutic intervention. The field of angiotensin biology has changed significantly over the last 50 years. Our original understanding of the crucial role of angiotensin II in the regulation of vascular tone and electrolyte homeostasis has been expanded to include the discovery of new angiotensins, their important role in cardiovascular inflammation and the development of clinically useful synthesis inhibitors and receptor antagonists. While less applied progress has been achieved in the kinin field, there are continuous discoveries in bradykinin physiology and in the complexity of kinin interactions with other proteins. The present review focuses on mechanisms and interactions of angiotensins and kinins that deal specifically with vascular endothelium.
Collapse
Affiliation(s)
- C Dimitropoulou
- Vascular Biology Center and Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, GA 30912-2500, USA
| | | | | | | | | |
Collapse
|
27
|
Pavar MC, Hanif K, Azam A, Lata S, Qadar Pasha MA, Pasha S. Structure–activity relationship study between Ornithyl-Proline and Lysyl-Proline based tripeptidomimics as angiotensin-converting enzyme inhibitors. Bioorg Med Chem Lett 2006; 16:2117-21. [PMID: 16464585 DOI: 10.1016/j.bmcl.2006.01.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Revised: 01/05/2006] [Accepted: 01/17/2006] [Indexed: 11/29/2022]
Abstract
A designed library of tripeptidomimics of Ornithyl-Proline (Orn-Pro) and Lysyl-Proline (Lys-Pro) conjugated with various unnatural amino acids and carboxylic acid derived heterocyclics was synthesized and screened for possible inhibitors of angiotensin-converting enzyme (ACE). Among the tripeptidomimics 10[MTP-Orn-Pro], 11[HTP-Orn-Pro], 14[TA-Orn-Pro] and 20[BPA-Orn-Pro] showed prominent inhibition with IC50 values in micromolar concentrations. Structure-activity relationship study indicated that C3 side chain of Orn as compared to C4 side chain of Lys at P1' position was better suited to inhibit ACE, with propionic acid (C3) derived heterocyclics and unnatural amino acids.
Collapse
Affiliation(s)
- Mahesh Chand Pavar
- Peptide Synthesis Lab, Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| | | | | | | | | | | |
Collapse
|
28
|
Arafat T, Awad R, Hamad M, Azzam R, Al-Nasan A, Jehanli A, Matalka K. Pharmacokinetics and pharmacodynamics profiles of enalapril maleate in healthy volunteers following determination of enalapril and enalaprilat by two specific enzyme immunoassays. J Clin Pharm Ther 2005; 30:319-28. [PMID: 15985045 DOI: 10.1111/j.1365-2710.2005.00646.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVES Most of the pharmacokinetic (PK) parameters for enalapril and enalaprilat were established following determination of the drug and its metabolite, using angiotensin converting enzyme (ACE) inhibition assays. In these methods, enalapril has to be hydrolysed to enalaprilat first and then assayed. The purpose of this study was to re-estimate the PK parameters of enalapril and enalaprilat in healthy volunteers using two specific enzyme immunoassays for enalapril and enalaprilat. METHODS The rate and extent of absorption of enalapril and enalaprilat from a 10-mg dose of two enalapril maleate commercial brands (Renetic and Enalapril) were estimated using a two-way-cross over design with 1-week washout period. Blood pressure was also measured at specified time intervals and correlated to enalaprilat plasma concentrations. RESULTS For enalapril, the AUC(o-->infinity) values (Mean+/-SD) were 450.0+/-199.5 and 479.6+/-215.6 ng h/mL, Cmax values were 313.5+/-139.6 and 310.1+/-186.6 ng/mL, Tmax values were 1.06+/-0.30 h and 1.13+/-0.22 h, and t1/2 ranged between 0.3 to 6.1 h (1.6+/-1.5) and 0.40 to 5.05 h (1.3+/-1.0), for the two brands. For enalaprilat, the AUC(o-->infinity) values were 266.9+/-122.7 and 255.9+/-121.8 ng h/ml, Cmax values were 54.8+/-29.5 and 57.2+/-29.0 ng/mL, Tmax values were 4.6+/-1.6 h and 4.3+/-1.45 h, and t1/2 ranged between 1.1 to 10.5 h (4.5+/-2.9) and 0.6 to 9.4 h (3.5+/-2.5) for the two brands. CONCLUSIONS Cmax values for enalapril are about 10 times those published in the literature and the rate and extent of absorption of the two brands of enalapril and their deesterification to enalaprilat following the administration of either brand were bioequivalent. Secondly, enalaprilat concentrations at 12-24 h following a single oral dose of enalapril in healthy volunteers were lower than those reported in the literature. The values reported here correlated with the return of blood pressure to predose level. Thirdly, enzyme immunoassays for enalapril and enalaprilat are better than ACE inhibition assays and can be used in bioequivalence assessment of enalapril and enalaprilat and for therapeutic drug monitoring in a clinical laboratory setting.
Collapse
Affiliation(s)
- T Arafat
- Faculty of Pharmacy and Medical Technology, University of Petra, Amman, Jordan.
| | | | | | | | | | | | | |
Collapse
|
29
|
Patchett AA. 2002 Alfred Burger Award Address in Medicinal Chemistry. Natural products and design: interrelated approaches in drug discovery. J Med Chem 2002; 45:5609-16. [PMID: 12477342 DOI: 10.1021/jm020424z] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Arthur A Patchett
- Department of Medicinal Chemistry, Merck Research Laboratories, Rahway, New Jersey 07065, USA.
| |
Collapse
|
30
|
Affiliation(s)
- J Menard
- Faculté de Médecine, Université Paris, 75270 Paris, France
| | | |
Collapse
|
31
|
Skellern GG. Recent advances in pharmaceutical chemistry--angiotensin-converting enzyme inhibitors. J Clin Pharm Ther 1989; 14:341-54. [PMID: 2684995 DOI: 10.1111/j.1365-2710.1989.tb00257.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- G G Skellern
- Department of Pharmacy, (Pharmaceutical Chemistry), University of Strathclyde, Glasgow, Scotland
| |
Collapse
|
32
|
Affiliation(s)
- J B Kostis
- Division of Cardiovascular Diseases & Hypertension, UMDNJ-Robert Wood Johnson Medical School, New Brunswick 08903-0019
| |
Collapse
|
33
|
Lant AF. Evolution of diuretics and ACE inhibitors, their renal and antihypertensive actions--parallels and contrasts. Br J Clin Pharmacol 1987; 23 Suppl 1:27S-41S. [PMID: 3034317 PMCID: PMC1386044 DOI: 10.1111/j.1365-2125.1987.tb03120.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The emergence of diuretic drugs and angiotensin converting enzyme (ACE) inhibitors ranks amongst the major therapeutic advances of modern medicine. The discovery of these drug groups arose largely by chance, yet each has dramatically influenced the treatment of congestive cardiac failure and arterial hypertension. The central role which diuretics have had in the management of both oedema and hypertension hinges on their ability to induce a net renal excretion of solute and water by selective interference with either active or passive ion transport processes in different segments of the nephron. Irrespective of sites of action, the continued antihypertensive action of diuretics is characterized by a reduction in plasma volume and extracellular fluid (ECF) volume that lasts for as long as the diuretic is given. The mechanism of this effect remains unclear but may involve autoregulatory reactions that leave cardiac output unaltered but maintain a sustained reduction in total peripheral resistance. ACE inhibitors also lower blood pressure by decreasing total peripheral resistance, leaving cardiac output, plasma volume and ECF volume unchanged. The detailed way these haemodynamic changes are achieved remains unknown but inhibition of converting enzyme present not only in the kidney but also in many extrarenal tissue sites, appears important. In both hypertension and cardiac failure, however, the kidney acts as a key target organ for ACE inhibitors. The increased renal vascular resistance and inappropriate renal salt excretion are reversed with enhanced renal blood flow and saluresis. Both angiotensin II (AII) and vasopressin-mediated contraction of glomerular mesangial cells is inhibited, making glomerular filtration more efficient. Reduced aldosterone secondary to blockade of AII formation contributes to saluresis whilst encouraging positive potassium balance. ACE inhibition also impairs breakdown of kinins which may contribute to intrarenal and peripheral vasodilation either on their own or via release of prostaglandins and other vasoactive substances. The hypotensive actions of diuretics are potentiated by ACE inhibition primarily through blockade of AII formation and prevention of secondary aldosteronism. In combination, these drugs permit low doses to be used because of their synergistic effects. Caution has to be exercised whenever ACE inhibition is used, without and especially with diuretics, in the management of renovascular hypertension and other low-perfusion states. In these circumstances, AII plays an important autoregulatory role in preserving glomerular filtration through an increase in post-glomerular resistance.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
|
34
|
Enalapril Maleate. ACTA ACUST UNITED AC 1987. [DOI: 10.1016/s0099-5428(08)60557-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
35
|
Wyvratt MJ, Patchett AA. Recent developments in the design of angiotensin-converting enzyme inhibitors. Med Res Rev 1985; 5:483-531. [PMID: 2999531 DOI: 10.1002/med.2610050405] [Citation(s) in RCA: 213] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Orally-active angiotensin-converting enzyme inhibitors are rapidly establishing themselves in the therapy of hypertension and congestive heart failure. Concerted efforts in a number of laboratories have now led to the discovery or synthesis of an unparalleled variety of potent inhibitors. The manner in which several of these inhibitors bind to ACE is beginning to be understood. It is hoped that some of the insights to be derived from the SAR and structural studies done with ACE inhibitors will be applicable to other enzyme targets as well. The success of ACE inhibitors as pharmacological tools and in the clinic will also quite certainly encourage future efforts to develop new enzyme inhibitor approaches to drug therapy.
Collapse
|