1
|
Torres-Herrero B, Armenia I, Ortiz C, de la Fuente JM, Betancor L, Grazú V. Opportunities for nanomaterials in enzyme therapy. J Control Release 2024; 372:619-647. [PMID: 38909702 DOI: 10.1016/j.jconrel.2024.06.035] [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] [Received: 03/10/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
In recent years, enzyme therapy strategies have rapidly evolved to catalyze essential biochemical reactions with therapeutic potential. These approaches hold particular promise in addressing rare genetic disorders, cancer treatment, neurodegenerative conditions, wound healing, inflammation management, and infectious disease control, among others. There are several primary reasons for the utilization of enzymes as therapeutics: their substrate specificity, their biological compatibility, and their ability to generate a high number of product molecules per enzyme unit. These features have encouraged their application in enzyme replacement therapy where the enzyme serves as the therapeutic agent to rectify abnormal metabolic and physiological processes, enzyme prodrug therapy where the enzyme initiates a clinical effect by activating prodrugs, and enzyme dynamic or starving therapy where the enzyme acts upon host substrate molecules. Currently, there are >20 commercialized products based on therapeutic enzymes, but approval rates are considerably lower than other biologicals. This has stimulated nanobiotechnology in the last years to develop nanoparticle-based solutions that integrate therapeutic enzymes. This approach aims to enhance stability, prevent rapid clearance, reduce immunogenicity, and even enable spatio-temporal activation of the therapeutic catalyst. This comprehensive review delves into emerging trends in the application of therapeutic enzymes, with a particular emphasis on the synergistic opportunities presented by incorporating enzymes into nanomaterials. Such integration holds the promise of enhancing existing therapies or even paving the way for innovative nanotherapeutic approaches.
Collapse
Affiliation(s)
- Beatriz Torres-Herrero
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Ilaria Armenia
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Cecilia Ortiz
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Jesús Martinez de la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Lorena Betancor
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Valeria Grazú
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
2
|
Hernández González LL, Pérez-Campos Mayoral L, Hernández-Huerta MT, Mayoral Andrade G, Martínez Cruz M, Ramos-Martínez E, Pérez-Campos Mayoral E, Cruz Hernández V, Antonio García I, Matias-Cervantes CA, Avendaño Villegas ME, Lastre Domínguez CM, Romero Díaz C, Ruiz-Rosado JDD, Pérez-Campos E. Targeting Neutrophil Extracellular Trap Formation: Exploring Promising Pharmacological Strategies for the Treatment of Preeclampsia. Pharmaceuticals (Basel) 2024; 17:605. [PMID: 38794175 PMCID: PMC11123764 DOI: 10.3390/ph17050605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Neutrophils, which constitute the most abundant leukocytes in human blood, emerge as crucial players in the induction of endothelial cell death and the modulation of endothelial cell responses under both physiological and pathological conditions. The hallmark of preeclampsia is endothelial dysfunction induced by systemic inflammation, in which neutrophils, particularly through the formation of neutrophil extracellular traps (NETs), play a pivotal role in the development and perpetuation of endothelial dysfunction and the hypertensive state. Considering the potential of numerous pharmaceutical agents to attenuate NET formation (NETosis) in preeclampsia, a comprehensive assessment of the extensively studied candidates becomes imperative. This review aims to identify mechanisms associated with the induction and negative regulation of NETs in the context of preeclampsia. We discuss potential drugs to modulate NETosis, such as NF-κβ inhibitors, vitamin D, and aspirin, and their association with mutagenicity and genotoxicity. Strong evidence supports the notion that molecules involved in the activation of NETs could serve as promising targets for the treatment of preeclampsia.
Collapse
Affiliation(s)
- Leticia Lorena Hernández González
- National Technology of Mexico/IT Oaxaca, Oaxaca de Juárez, Oaxaca 68030, Mexico; (L.L.H.G.); (M.M.C.); (C.M.L.D.); (C.R.D.)
- Faculty of Biological Systems and Technological Innovation, Autonomous University “Benito Juárez” of Oaxaca, Oaxaca 68125, Mexico
| | - Laura Pérez-Campos Mayoral
- Research Center, Faculty of Medicine UNAM-UABJO, Autonomous University “Benito Juárez” of Oaxaca (UABJO), Oaxaca 68020, Mexico; (L.P.-C.M.); (G.M.A.); (E.P.-C.M.)
| | - María Teresa Hernández-Huerta
- CONAHCyT, Faculty of Medicine and Surgery, Autonomous University “Benito Juárez” of Oaxaca (UABJO), Oaxaca 68020, Mexico; (M.T.H.-H.); (C.A.M.-C.)
| | - Gabriel Mayoral Andrade
- Research Center, Faculty of Medicine UNAM-UABJO, Autonomous University “Benito Juárez” of Oaxaca (UABJO), Oaxaca 68020, Mexico; (L.P.-C.M.); (G.M.A.); (E.P.-C.M.)
| | - Margarito Martínez Cruz
- National Technology of Mexico/IT Oaxaca, Oaxaca de Juárez, Oaxaca 68030, Mexico; (L.L.H.G.); (M.M.C.); (C.M.L.D.); (C.R.D.)
| | - Edgar Ramos-Martínez
- School of Sciences, Autonomous University “Benito Juárez” of Oaxaca (UABJO), Oaxaca 68020, Mexico;
| | - Eduardo Pérez-Campos Mayoral
- Research Center, Faculty of Medicine UNAM-UABJO, Autonomous University “Benito Juárez” of Oaxaca (UABJO), Oaxaca 68020, Mexico; (L.P.-C.M.); (G.M.A.); (E.P.-C.M.)
| | | | | | - Carlos Alberto Matias-Cervantes
- CONAHCyT, Faculty of Medicine and Surgery, Autonomous University “Benito Juárez” of Oaxaca (UABJO), Oaxaca 68020, Mexico; (M.T.H.-H.); (C.A.M.-C.)
| | - Miriam Emily Avendaño Villegas
- National Technology of Mexico/IT Oaxaca, Oaxaca de Juárez, Oaxaca 68030, Mexico; (L.L.H.G.); (M.M.C.); (C.M.L.D.); (C.R.D.)
| | | | - Carlos Romero Díaz
- National Technology of Mexico/IT Oaxaca, Oaxaca de Juárez, Oaxaca 68030, Mexico; (L.L.H.G.); (M.M.C.); (C.M.L.D.); (C.R.D.)
- Research Center, Faculty of Medicine UNAM-UABJO, Autonomous University “Benito Juárez” of Oaxaca (UABJO), Oaxaca 68020, Mexico; (L.P.-C.M.); (G.M.A.); (E.P.-C.M.)
| | - Juan de Dios Ruiz-Rosado
- Kidney and Urinary Tract Research Center, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Eduardo Pérez-Campos
- National Technology of Mexico/IT Oaxaca, Oaxaca de Juárez, Oaxaca 68030, Mexico; (L.L.H.G.); (M.M.C.); (C.M.L.D.); (C.R.D.)
- Clinical Pathology Laboratory, “Eduardo Pérez Ortega”, Oaxaca 68000, Mexico
| |
Collapse
|
3
|
Zeng X, Liu Y, Fan Y, Wu D, Meng Y, Qin M. Agents for the Treatment of Gout: Current Advances and Future Perspectives. J Med Chem 2023; 66:14474-14493. [PMID: 37908076 DOI: 10.1021/acs.jmedchem.3c01710] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Gout is characterized by hyperuricemia and the deposition of monosodium urate (MSU) crystals around joints. Despite the availability of several drugs on the market, its treatment remains challenging owing to the notable side effects, such as hepatorenal toxicity and cardiovascular complications, that are associated with most existing agents. This perspective aims to summarize the current research progress in the development of antigout agents, particularly focusing on xanthine oxidase (XO) and urate anion transporter 1 (URAT1) inhibitors from a medicinal chemistry viewpoint and their preliminary structure-activity relationships (SARs). This perspective provides valuable insights and theoretical guidance to medicinal chemists for the discovery of antigout agents with novel chemical structures, better efficiency, and lower toxicity.
Collapse
Affiliation(s)
- Xiaoyi Zeng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yajing Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yuxin Fan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Di Wu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yangyang Meng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Mingze Qin
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| |
Collapse
|
4
|
Peng X, Li X, Xie B, Lai Y, Sosnik A, Boucetta H, Chen Z, He W. Gout therapeutics and drug delivery. J Control Release 2023; 362:728-754. [PMID: 37690697 DOI: 10.1016/j.jconrel.2023.09.011] [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] [Received: 05/28/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
Gout is a common inflammatory arthritis caused by persistently elevated uric acid levels. With the improvement of people's living standards, the consumption of processed food and the widespread use of drugs that induce elevated uric acid, gout rates are increasing, seriously affecting the human quality of life, and becoming a burden to health systems worldwide. Since the pathological mechanism of gout has been elucidated, there are relatively effective drug treatments in clinical practice. However, due to (bio)pharmaceutical shortcomings of these drugs, such as poor chemical stability and limited ability to target the pathophysiological pathways, traditional drug treatment strategies show low efficacy and safety. In this scenario, drug delivery systems (DDS) design that overcome these drawbacks is urgently called for. In this review, we initially describe the pathological features, the therapeutic targets, and the drugs currently in clinical use and under investigation to treat gout. We also comprehensively summarize recent research efforts utilizing lipid, polymeric and inorganic carriers to develop advanced DDS for improved gout management and therapy.
Collapse
Affiliation(s)
- Xiuju Peng
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Xiaotong Li
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Bing Xie
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Yaoyao Lai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Alejandro Sosnik
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Hamza Boucetta
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| |
Collapse
|
5
|
Simay S, Akbarzadeh-Khiavi M, Pourseif MM, Barar J, Safary A, Omidi Y. Recombinant production and characterization of L-glutaminase (glsA) as a promiscuity therapeutic enzyme. Appl Microbiol Biotechnol 2022; 106:5511-5524. [PMID: 35876873 DOI: 10.1007/s00253-022-12058-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 06/14/2022] [Accepted: 06/28/2022] [Indexed: 11/02/2022]
Abstract
Because of the therapeutical impacts of hydrolytic enzymes in different diseases, in particular malignancies, we aimed to produce a recombinant putative L-glutaminase (GLS ASL-1) from a recently characterized halo-thermotolerant Bacillus sp. SL-1. For this purpose, the glsA gene was identified and efficiently overexpressed in the Origami™ B (DE3) strain. The yield of the purified GLS ASL-1 was ~ 20 mg/L, indicating a significant expression of recombinant enzyme in the Origami. The enzyme activity assay revealed a significant hydrolytic effect of the recombinant GLS ASL-1 on L-asparagine (Asn) (i.e., Km 39.8 μM, kcat 19.9 S-1) with a minimal affinity for L-glutamine (Gln). The GLS ASL-1 significantly suppressed the growth of leukemic Jurkat cells through apoptosis induction (47.5%) in the IC50 dosage of the enzyme. The GLS ASL-1 could also change the Bax/Bcl2 expression ratio, indicating its apoptotic effect on cancer cells. The in silico analysis was conducted to predict structural features related to the histidine-tag exposure in the N- or C-terminal of the recombinant GLS ASL-1. In addition, molecular docking simulation for substrate specificity revealed a greater binding affinity of Asn to the enzyme binding-site residues than Gln, which was confirmed in experimental procedures as well. In conclusion, the current study introduced a recombinant GLS ASL-1 with unique functional and structural features, highlighting its potential pharmaceutical and medical importance. GLS ASL-1 represents the first annotated enzyme from Bacillus with prominent asparaginase activity, which can be considered for developing alternative enzymes in therapeutic applications. KEY POINTS: • Hydrolytic enzymes have critical applications in different types of human malignancies. • A recombinant L-glutaminase (GLS ASL-1) was produced from halo-thermotolerant Bacillus sp. SL-1. • GLS ASL-1 displayed a marked hydrolytic activity on L-asparagine compared to the L-glutamine. • GLS ASL-1 with significant substrate promiscuity may be an alternative for developing novel pharmaceuticals.
Collapse
Affiliation(s)
- Shayan Simay
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Akbarzadeh-Khiavi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad M Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azam Safary
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328, USA.
| |
Collapse
|
6
|
Vanderstocken G, Woolf NL, Trigiante G, Jackson J, McGoldrick R. Harnessing the Potential of Enzymes as Inhaled Therapeutics in Respiratory Tract Diseases: A Review of the Literature. Biomedicines 2022; 10:biomedicines10061440. [PMID: 35740461 PMCID: PMC9220205 DOI: 10.3390/biomedicines10061440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/01/2022] [Accepted: 06/14/2022] [Indexed: 11/24/2022] Open
Abstract
Respiratory tract diseases (RTDs) are a global cause of mortality and affect patient well-being and quality of life. Specifically, there is a high unmet need concerning respiratory tract infections (RTIs) due to limitations of vaccines and increased antibiotic resistance. Enzyme therapeutics, and in particular plant-based enzymes, represent an underutilised resource in drug development warranting further attention. This literature review aims to summarise the current state of enzyme therapeutics in medical applications, with a focus on their potential to improve outcomes in RTDs, including RTIs. We used a narrative review approach, searching PubMed and clinicaltrials.gov with search terms including: enzyme therapeutics, enzyme therapy, inhaled therapeutics, botanical enzyme therapeutics, plant enzymes, and herbal extracts. Here, we discuss the advantages and challenges of enzyme therapeutics in the setting of RTDs and identify and describe several enzyme therapeutics currently used in the respiratory field. In addition, the review includes recent developments concerning enzyme therapies and plant enzymes in (pre-)clinical stages. The global coronavirus disease 2019 (COVID-19) pandemic has sparked development of several promising new enzyme therapeutics for use in the respiratory setting, and therefore, it is timely to provide a summary of recent developments, particularly as these therapeutics may also prove beneficial in other RTDs.
Collapse
Affiliation(s)
| | - Nicholas L. Woolf
- Inspira Pharmaceuticals Limited, 27 Old Gloucester Street, London WC1N 3AX, UK; (N.L.W.); (J.J.)
| | - Giuseppe Trigiante
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 2AT, UK;
| | - Jessica Jackson
- Inspira Pharmaceuticals Limited, 27 Old Gloucester Street, London WC1N 3AX, UK; (N.L.W.); (J.J.)
| | - Rory McGoldrick
- Inspira Pharmaceuticals Limited, 27 Old Gloucester Street, London WC1N 3AX, UK; (N.L.W.); (J.J.)
- Correspondence:
| |
Collapse
|
7
|
Ding M, Zhang Y, Li J, Pu K. Bioenzyme-based nanomedicines for enhanced cancer therapy. NANO CONVERGENCE 2022; 9:7. [PMID: 35119544 PMCID: PMC8816986 DOI: 10.1186/s40580-022-00297-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/04/2022] [Indexed: 05/09/2023]
Abstract
Bioenzymes that catalyze reactions within living systems show a great promise for cancer therapy, particularly when they are integrated with nanoparticles to improve their accumulation into tumor sites. Nanomedicines can deliver toxic bioenzymes into cancer cells to directly cause their death for cancer treatment. By modulating the tumor microenvironment, such as pH, glucose concentration, hypoxia, redox levels and heat shock protein expression, bioenzyme-based nanomedicines play crucial roles in improving the therapeutic efficacy of treatments. Moreover, bioenzyme-mediated degradation of the major components in tumor extracellular matrix greatly increases the penetration and retention of nanoparticles in deep tumors and infiltration of immune cells into tumor tissues, thus enhancing the efficacies of chemotherapy, phototherapy and immunotherapy. In this review, we summarize the recent progresses of bioenzyme-based nanomedicines for enhanced cancer therapy. The design and working mechanisms of the bioenzyme-based nanomedicines to achieve enhanced chemotherapy, photothermal therapy, photodynamic therapy, chemodynamic therapy, radiotherapy and immunotherapy are introduced in detail. At the end of this review, a conclusion and current challenges and perspectives in this field are given.
Collapse
Affiliation(s)
- Mengbin Ding
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Yijing Zhang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Jingchao Li
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China.
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
| |
Collapse
|
8
|
Lee MJ, Lee I, Wang K. Recent Advances in RNA Therapy and Its Carriers to Treat the Single-Gene Neurological Disorders. Biomedicines 2022; 10:biomedicines10010158. [PMID: 35052837 PMCID: PMC8773368 DOI: 10.3390/biomedicines10010158] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
The development of new sequencing technologies in the post-genomic era has accelerated the identification of causative mutations of several single gene disorders. Advances in cell and animal models provide insights into the underlining pathogenesis, which facilitates the development and maturation of new treatment strategies. The progress in biochemistry and molecular biology has established a new class of therapeutics—the short RNAs and expressible long RNAs. The sequences of therapeutic RNAs can be optimized to enhance their stability and translatability with reduced immunogenicity. The chemically-modified RNAs can also increase their stability during intracellular trafficking. In addition, the development of safe and high efficiency carriers that preserves the integrity of therapeutic RNA molecules also accelerates the transition of RNA therapeutics into the clinic. For example, for diseases that are caused by genetic defects in a specific protein, an effective approach termed “protein replacement therapy” can provide treatment through the delivery of modified translatable mRNAs. Short interference RNAs can also be used to treat diseases caused by gain of function mutations or restore the splicing aberration defects. Here we review the applications of newly developed RNA-based therapeutics and its delivery and discuss the clinical evidence supporting the potential of RNA-based therapy in single-gene neurological disorders.
Collapse
Affiliation(s)
- Ming-Jen Lee
- Department of Neurology, National Taiwan University Hospital, Taipei 10012, Taiwan;
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 10012, Taiwan
| | - Inyoul Lee
- Institute for Systems Biology, Seattle, WA 98109, USA;
| | - Kai Wang
- Institute for Systems Biology, Seattle, WA 98109, USA;
- Correspondence: ; Tel.: +1-206-732-1336
| |
Collapse
|
9
|
Parveen A, Devika R. Fibrinolytic Enzyme - An Overview. Curr Pharm Biotechnol 2022; 23:1336-1345. [PMID: 34983344 DOI: 10.2174/1389201023666220104143113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/21/2021] [Accepted: 10/13/2021] [Indexed: 11/22/2022]
Abstract
Cardiovascular diseases, like coronary heart disease or artery disorders (arteriosclerosis, including artery solidification), heart failure (myocardial infarction), arrhythmias, congestive heart condition, stroke, elevated vital signs (hypertension), rheumatic heart disorder, and other circulatory system dysfunctions are the most common causes of death worldwide. Cardiovascular disorders are treated with stenting, coronary bypass surgery grafting, anticoagulants, antiplatelet agents, and other pharmacological and surgical procedures; however, these have limitations due to their adverse effects. Fibrinolytic agents degrade fibrin through enzymatic and biochemical processes. There are various enzymes that are currently used as a treatment for CVDs, like Streptokinase, Nattokinase, Staphylokinase, Urokinase, etc. These enzymes are derived from various sources like bacteria, fungi, algae, marine organisms, plants, snakes, and other organisms. This review deals with the fibrinolytic enzymes, their mechanisms, sources, and their therapeutic potential.
Collapse
Affiliation(s)
- Parveen A
- Department of Biotechnology, Biotechnology, Aarupadai Institute of Technology, Vinayaka Missions University, Chennai, India
| | - Devika R
- Department of Biotechnology, Biotechnology, Aarupadai Institute of Technology, Vinayaka Missions University, Chennai, India
| |
Collapse
|
10
|
Hennigan JN, Lynch MD. The past, present, and future of enzyme-based therapies. Drug Discov Today 2022; 27:117-133. [PMID: 34537332 PMCID: PMC8714691 DOI: 10.1016/j.drudis.2021.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/15/2021] [Accepted: 09/10/2021] [Indexed: 01/03/2023]
Abstract
Enzyme-based therapeutics (EBTs) have the potential to tap into an almost unmeasurable amount of enzyme biodiversity and treat myriad conditions. Although EBTs were some of the first biologics used clinically, the rate of development of newer EBTs has lagged behind that of other biologics. Here, we review the history of EBTs, and discuss the state of each class of EBT, their potential clinical advantages, and the unique challenges to their development. Additionally, we discuss key remaining technical barriers that, if addressed, could increase the diversity and rate of the development of EBTs.
Collapse
Affiliation(s)
| | - Michael D Lynch
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
11
|
Pyrgidis N, Vakalopoulos I, Sountoulides P. Endocrine consequences of treatment with the new androgen receptor axis-targeted agents for advanced prostate cancer. Hormones (Athens) 2021; 20:73-84. [PMID: 33140306 DOI: 10.1007/s42000-020-00251-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/08/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE Prostate cancer (PCa) is the commonest non-cutaneous malignancy worldwide and the second cause of cancer death among males in the USA. Approval of the new androgen receptor axis-targeted (ARAT) agents (abiraterone acetate, enzalutamide, apalutamide, and darolutamide) has altered the course of advanced PCa. We aimed to assess the endocrine and metabolic adverse events associated with treatment using ARAT compounds. METHODS We searched the PubMed, Cochrane Library, and Scopus databases from database inception to August 2020. We included randomized controlled trials reporting the endocrine and metabolic side effects of ARAT agents compared to each other or to placebo. RESULTS Although metastatic PCa remains incurable, ARAT medications combined with androgen deprivation therapy improve overall metastasis-free and progression-free survival in metastatic hormone-sensitive PCa, non-metastatic castration-resistant PCa, and metastatic castration-resistant PCa patients. This benefit comes at the cost of certain endocrine and metabolic consequences. Treatment with abiraterone acetate induces mineralocorticoid excess, hypokalemia, hypertension, elevated liver function tests, insulin resistance, and hyperglycemia. Enzalutamide may induce or worsen hypertension and increase the risk of falls and fractures in elderly patients, while common endocrine adverse events of apalutamide include hypothyroidism, hypertension, and skin rash. On the other hand, darolutamide seems to have a somewhat safer endocrine and metabolic profile. CONCLUSION Treatment of advanced PCa should be personalized, with administration of a combination of androgen deprivation therapy, ARAT agents, and chemotherapy being based on the patient's safety profile and the risk of side effects.
Collapse
Affiliation(s)
- Nikolaos Pyrgidis
- 1st Department of Urology, Aristotle University of Thessaloniki, 15-17 Agiou Evgeniou Street, TK 55133, Thessaloniki, Greece
| | - Ioannis Vakalopoulos
- 1st Department of Urology, Aristotle University of Thessaloniki, 15-17 Agiou Evgeniou Street, TK 55133, Thessaloniki, Greece
| | - Petros Sountoulides
- 1st Department of Urology, Aristotle University of Thessaloniki, 15-17 Agiou Evgeniou Street, TK 55133, Thessaloniki, Greece.
| |
Collapse
|
12
|
Losada-Garcia N, Jimenez-Alesanco A, Velazquez-Campoy A, Abian O, Palomo JM. Enzyme/Nanocopper Hybrid Nanozymes: Modulating Enzyme-like Activity by the Protein Structure for Biosensing and Tumor Catalytic Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:5111-5124. [PMID: 33472360 PMCID: PMC8486171 DOI: 10.1021/acsami.0c20501] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/12/2021] [Indexed: 05/30/2023]
Abstract
Artificial enzymes with modulated enzyme-mimicking activities of natural systems represent a challenge in catalytic applications. Here, we show the creation of artificial Cu metalloenzymes based on the generation of Cu nanoparticles in an enzyme matrix. Different enzymes were used, and the structural differences between the enzymes especially influenced the controlled the size of the nanoparticles and the environment that surrounds them. Herein, we demonstrated that the oxidase-like catalytic activity of these copper nanozymes was rationally modulated by enzyme used as a scaffold, with a special role in the nanoparticle size and their environment. In this sense, these nanocopper hybrids have confirmed the ability to mimic a unique enzymatic activity completely different from the natural activity of the enzyme used as a scaffold, such as tyrosinase-like activity or as Fenton catalyst, which has extremely higher stability than natural mushroom tyrosinase. More interestingly, the oxidoreductase-like activity of nanocopper hybrids was cooperatively modulated with the synergistic effect between the enzyme and the nanoparticles improving the catalase activity (no peroxidase activity). Additionally, a novel dual (metallic and enzymatic activity) of the nanozyme made the highly improved catechol-like activity interesting for the design of 3,4-dihydroxy-l-phenylalanine (l-DOPA) biosensor for detection of tyrosinase. These hybrids also showed cytotoxic activity against different tumor cells, interesting in biocatalytic tumor therapy.
Collapse
Affiliation(s)
- Noelia Losada-Garcia
- Department
of Biocatalysis, Institute of Catalysis
(CSIC), c/Marie curie 2, Cantoblanco Campus UAM, 28049 Madrid, Spain
| | - Ana Jimenez-Alesanco
- Instituto
de Biocomputación y Física de Sistemas Complejos, Joint
Units IQFR-CSIC-BIFI, and GBsC-CSIC-BIFI, Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Adrian Velazquez-Campoy
- Fundación
ARAID, Gobierno de Aragón, 50018 Zaragoza, Spain
- Instituto
de Biocomputación y Física de Sistemas Complejos, Joint
Units IQFR-CSIC-BIFI, and GBsC-CSIC-BIFI, Universidad de Zaragoza, 50009 Zaragoza, Spain
- Fundación
Instituto de Investigación Sanitaria de Aragón (IIS
Aragón), 50009 Zaragoza, Spain
- Centro
de Investigación Biomédica en Red en el Área
Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
- Departamento
de Bioquímica y Biología Molecular y Celular, Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Olga Abian
- Instituto
de Biocomputación y Física de Sistemas Complejos, Joint
Units IQFR-CSIC-BIFI, and GBsC-CSIC-BIFI, Universidad de Zaragoza, 50009 Zaragoza, Spain
- Fundación
Instituto de Investigación Sanitaria de Aragón (IIS
Aragón), 50009 Zaragoza, Spain
- Centro
de Investigación Biomédica en Red en el Área
Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
- Departamento
de Bioquímica y Biología Molecular y Celular, Universidad de Zaragoza, 50009 Zaragoza, Spain
- Instituto
Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
| | - Jose M. Palomo
- Department
of Biocatalysis, Institute of Catalysis
(CSIC), c/Marie curie 2, Cantoblanco Campus UAM, 28049 Madrid, Spain
| |
Collapse
|
13
|
Í Dali C, Groeschel S, Moldovan M, Farah MH, Krägeloh-Mann I, Wasilewski M, Li J, Barton N, Krarup C. Intravenous arylsulfatase A in metachromatic leukodystrophy: a phase 1/2 study. Ann Clin Transl Neurol 2020; 8:66-80. [PMID: 33332761 PMCID: PMC7818087 DOI: 10.1002/acn3.51254] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/12/2020] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Metachromatic leukodystrophy (MLD) is an autosomal recessive lysosomal storage disease caused by deficient activity of arylsulfatase A (ASA), resulting in severe motor and cognitive dysfunction. This phase 1/2 study evaluated the safety and efficacy of intravenous (IV) recombinant human ASA (rhASA; HGT-1111, previously known as Metazym) in children with MLD. METHODS Thirteen children with MLD (symptom onset < 4 years of age) were enrolled in an open-label, nonrandomized, dose-escalation trial and received IV rhASA at 50, 100, or 200 U/kg body weight every 14 (± 4) days for 52 weeks (NCT00418561; NCT00633139). Eleven children continued to receive rhASA at 100 or 200 U/kg during a 24-month extension period (NCT00681811). Outcome measures included safety observations, changes in motor and cognitive function, and changes in nerve conduction and morphometry. RESULTS There were no serious adverse events considered related to IV rhASA. Motor function and developmental testing scores declined during the study in all dose groups; no significant differences were observed between groups. Nerve conduction studies and morphometric analysis indicated that peripheral nerve pathology did not worsen during the study in any dose group. INTERPRETATION IV rhASA was generally well tolerated. There was no evidence of efficacy in preventing motor and cognitive deterioration, suggesting that IV rhASA may not cross the blood-brain barrier in therapeutic quantities. The relative stability of peripheral nerve function during the study indicates that rhASA may be beneficial if delivered to the appropriate target site and supports the development of rhASA for intrathecal administration in MLD.
Collapse
Affiliation(s)
- Christine Í Dali
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Samuel Groeschel
- Department of Neuropediatrics, University Children's Hospital Tübingen, Tübingen, Germany
| | - Mihai Moldovan
- Department of Clinical Neurophysiology, Rigshospitalet, Copenhagen, Denmark.,Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Mohamed H Farah
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ingeborg Krägeloh-Mann
- Department of Neuropediatrics, University Children's Hospital Tübingen, Tübingen, Germany
| | - Margaret Wasilewski
- Shire (a member of the Takeda group of companies), Lexington, Massachusetts, USA
| | - Jing Li
- Shire (a member of the Takeda group of companies), Lexington, Massachusetts, USA
| | - Norman Barton
- Shire (a member of the Takeda group of companies), Lexington, Massachusetts, USA
| | - Christian Krarup
- Department of Clinical Neurophysiology, Rigshospitalet, Copenhagen, Denmark.,Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Wang F, Moen DR, Sauni C, Kan SH, Li S, Le SQ, Lomenick B, Zhang X, Ekins S, Singamsetty S, Wood J, Dickson PI, Chou TF. Enzyme Replacement Therapy for Mucopolysaccharidosis IIID using Recombinant Human α- N-Acetylglucosamine-6-Sulfatase in Neonatal Mice. Mol Pharm 2020; 18:214-227. [PMID: 33320673 DOI: 10.1021/acs.molpharmaceut.0c00831] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There is currently no cure or effective treatment available for mucopolysaccharidosis type IIID (MPS IIID, Sanfilippo syndrome type D), a lysosomal storage disorder (LSD) caused by the deficiency of α-N-acetylglucosamine-6-sulfatase (GNS). The clinical symptoms of MPS IIID, like other subtypes of Sanfilippo syndrome, are largely localized to the central nervous system (CNS), and any treatments aiming to ameliorate or reverse the catastrophic and fatal neurologic decline caused by this disease need to be delivered across the blood-brain barrier. Here, we report a proof-of-concept enzyme replacement therapy (ERT) for MPS IIID using recombinant human α-N-acetylglucosamine-6-sulfatase (rhGNS) via intracerebroventricular (ICV) delivery in a neonatal MPS IIID mouse model. We overexpressed and purified rhGNS from CHO cells with a specific activity of 3.9 × 104 units/mg protein and a maximal enzymatic activity at lysosomal pH (pH 5.6), which was stable for over one month at 4 °C in artificial cerebrospinal fluid (CSF). We demonstrated that rhGNS was taken up by MPS IIID patient fibroblasts via the mannose 6-phosphate (M6P) receptor and reduced intracellular glycosaminoglycans to normal levels. The delivery of 5 μg of rhGNS into the lateral cerebral ventricle of neonatal MPS IIID mice resulted in normalization of the enzymatic activity in brain tissues; rhGNS was found to be enriched in lysosomes in MPS IIID-treated mice relative to the control. Furthermore, a single dose of rhGNS was able to reduce the accumulated heparan sulfate and β-hexosaminidase. Our results demonstrate that rhGNS delivered into CSF is a potential therapeutic option for MPS IIID that is worthy of further development.
Collapse
Affiliation(s)
- Feng Wang
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Derek R Moen
- Phoenix Nest Inc., Brooklyn, New York 11232, United States
| | - Chelsee Sauni
- Phoenix Nest Inc., Brooklyn, New York 11232, United States
| | - Shih-Hsin Kan
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Research Administration, CHOC Children's Hospital, Orange, California 92868, United States
| | - Shan Li
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Steven Q Le
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Brett Lomenick
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, California 91125, United States
| | - Xiaoyi Zhang
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States
| | - Sean Ekins
- Phoenix Nest Inc., Brooklyn, New York 11232, United States
| | | | - Jill Wood
- Phoenix Nest Inc., Brooklyn, New York 11232, United States
| | - Patricia I Dickson
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Tsui-Fen Chou
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, California 90502, United States.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States.,Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
15
|
LC-MS/MS assays to quantify sulfatides and lysosulfatide in cerebrospinal fluid of metachromatic leukodystrophy patients. Bioanalysis 2020; 12:1621-1633. [PMID: 33151743 DOI: 10.4155/bio-2020-0200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Two separate LC-MS/MS assays were developed to quantitate sulfatides and lysosulfatide in human cerebrospinal fluid (CSF). Materials & methods: Lysosulfatide and the 15 most abundant sulfatide species were quantitated by LC-MS/MS using artificial CSF as surrogate matrix to prepare calibration curves. Results: Validation criteria were met (linear range: 0.02-1.00 μg/ml sulfatides [0.02-1.00 ng/ml lysosulfatide]); accuracy/precision were within ±15%. CSF from 21 children with metachromatic leukodystrophy had significantly higher sulfatide and lysosulfatide concentrations than CSF from 60 healthy children (p < 0.0001). Worse motor function correlated with higher CSF sulfatide (p = 0.0087) and lysosulfatide (p = 0.0034) levels. Conclusion: These assays, validated in patients with metachromatic leukodystrophy, may aid the clinical assessment of therapeutic responses.
Collapse
|
16
|
Í Dali C, Sevin C, Krägeloh-Mann I, Giugliani R, Sakai N, Wu J, Wasilewski M. Safety of intrathecal delivery of recombinant human arylsulfatase A in children with metachromatic leukodystrophy: Results from a phase 1/2 clinical trial. Mol Genet Metab 2020; 131:235-244. [PMID: 32792226 DOI: 10.1016/j.ymgme.2020.07.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Metachromatic leukodystrophy (MLD) is an autosomal recessive disorder caused by deficient arylsulfatase A (ASA) activity and characterized by neurological involvement that results in severe disability and premature death. We examined the safety and tolerability of intrathecally delivered recombinant human ASA (rhASA; SHP611, now TAK-611) in children with MLD (NCT01510028). Secondary endpoints included change in cerebrospinal fluid (CSF) sulfatide and lysosulfatide levels, and motor function (assessed by Gross Motor Function Measure-88 total score). METHODS Twenty-four children with MLD who experienced symptom onset aged ≤ 30 months were enrolled. Patients received rhASA every other week (EOW) for 38 weeks at 10, 30, or 100 mg (cohorts 1-3; n = 6 per cohort), or 100 mg manufactured using a revised process (cohort 4; n = 6). RESULTS No rhASA-related serious adverse events (SAEs) were observed; 25% of patients experienced an SAE related to the intrathecal device or drug delivery method. Mean CSF sulfatide and lysosulfatide levels fell to within normal ranges in both 100 mg cohorts following treatment. Although there was a general decline in motor function over time, there was a tendency towards a less pronounced decline in patients receiving 100 mg. CONCLUSION Intrathecal rhASA was generally well tolerated at doses up to 100 mg EOW. These preliminary data support further development of rhASA as a therapy for patients with MLD.
Collapse
Affiliation(s)
- Christine Í Dali
- Department of Clinical Genetics, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.
| | | | - Ingeborg Krägeloh-Mann
- Department of Neuropediatrics, University Children's Hospital Tübingen, Tübingen, Germany.
| | - Roberto Giugliani
- Medical Genetics Service, HCPA, Department of Genetics, UFRGS, and INAGEMP, Porto Alegre, Brazil.
| | | | - James Wu
- Shire, a member of the Takeda group of companies, Lexington, MA, USA.
| | | |
Collapse
|
17
|
Politei J. Fabry disease during the COVID-19 pandemic. Why and how treatment should be continued. Mol Genet Metab 2020; 130:227-229. [PMID: 32561366 PMCID: PMC7274950 DOI: 10.1016/j.ymgme.2020.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 11/29/2022]
Abstract
Fabry disease is an X-linked disease due to a deficiency of the lysosomal enzyme alpha-galactosidase A. Clinical symptoms in classically affected males include acroparesthesia, anhydrosis and angiokeratoma, which may present during childhood followed by cardiac, cerebral and renal complications. Even though pulmonary involvement is not widely appreciated by clinicians, an obstructive lung disease is another recognized component of Fabry disease. Coronavirus Disease-19 (COVID-19), caused by the SARS-CoV-2 virus was labeled as a global pandemic and patients with Fabry disease can be considered at high risk of developing severe complications. The impact of COVID-19 on patients with Fabry disease receiving enzyme replacement therapy is still unknown. Many patients who receive treatment in the hospital experienced infusion disruptions due to fear of infection. Effects of temporary treatment interruption was described in more detail in other lysosomal storage diseases, but the recommencement of therapy does not fully reverse clinical decline due to the temporary discontinuation. When possible, home-therapy seems to be the most efficient way to maintain enzyme replacement therapy access during pandemic. Sentence take-home message: Home-therapy, when possible, seems to be the most efficient way to maintain enzyme replacement therapy access during pandemic in patients with Fabry disease.
Collapse
Affiliation(s)
- Juan Politei
- Fundation for the Study of Neurometabolic Diseases, FESEN, Argentina.
| |
Collapse
|
18
|
van Doorn J. Insulin-like growth factor-II and bioactive proteins containing a part of the E-domain of pro-insulin-like growth factor-II. Biofactors 2020; 46:563-578. [PMID: 32026557 PMCID: PMC7497164 DOI: 10.1002/biof.1623] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022]
Abstract
Insulin-like growth factor (IGF)-II is considered to function as an important fetal growth factor, which is structurally and functionally related to IGF-I and proinsulin. At least in vitro, IGF-II actions are mediated through the IGF-I receptor and to a lesser extent the insulin receptor. After birth, the function of IGF-II is less clear although in adults the serum level of IGF-II exceeds that of IGF-I several fold. The IGF-II gene is maternally imprinted, with exception of the liver and several parts of the brain, where it is expressed from both alleles. The regulation, organization, and translation of the IGF-II gene is complex, with five different putative promotors leading to a range of noncoding and coding mRNAs. The 180-amino acid pre-pro-IGF-II translation product can be divided into five domains and include a N-terminal signal peptide of 24 amino acid residues, the 67 amino acid long mature protein, and an 89 residues extension at the COOH terminus, designated as the E-domain. After removal of the signal peptide, the processing of pro-IGF-II into mature IGF-II requires various steps including glycosylation of the E-domain followed by the action of endo-proteases. Several of these processing intermediates can be found in the human circulation. There is increasing evidence that, besides IGF-II, several incompletely processed precursor forms of the protein, and even a 34-amino acid peptide (preptin) derived from the E-domain of pro-IGF-II, exhibit distinct biological activities. This review will focus on the current insights regarding the specific roles of the latter proteins in cancer, glucose homeostasis, and bone physiology. To address this topic clearly in the right context, a concise overview of the biological and biochemical properties of IGF-II and several relevant aspects of the IGF system will be provided.
Collapse
Affiliation(s)
- Jaap van Doorn
- Department of Genetics, Section Metabolic DiagnosticsUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
19
|
Madrakhimov SB, Yang JY, Ahn DH, Han JW, Ha TH, Park TK. Peripapillary Intravitreal Injection Improves AAV-Mediated Retinal Transduction. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:647-656. [PMID: 32300611 PMCID: PMC7152690 DOI: 10.1016/j.omtm.2020.03.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/23/2020] [Indexed: 12/31/2022]
Abstract
The intravitreal (IVT) injection method is a choice when targeting the inner retina for gene therapy. However, the transduction efficiency of adeno-associated virus (AAV) vectors administered by the IVT route is usually low and may be affected by several factors. To improve the transduction efficiency, we developed a novel illuminated long-needle attached injection system and injected AAV2-CMV (cytomegalovirus)-EGFP in front of the retina in rabbit eyes. Ophthalmological examinations were performed and the levels of pro-inflammatory cytokines in the aqueous humor were assessed at the baseline and 1 month, and the results were compared with those of the conventional injection method. Retinal tissues were used for immunohistochemistry. In the ophthalmological examinations, no significant inflammatory signs were detected in both groups, except for transient, mild hyperemia. In the tissues of the rabbits in the peripapillary injection group, significantly increased GFP expression was detected at the ganglion cell and the inner nuclear layers (p < 0.01). There were no differences between groups in glial activation and expressions of interleukin (IL)-6 and IL-8. These results suggest that peripapillary IVT injection in front of the retina would be safe and efficiently transduce viral vectors into the retina of large animals and is considered as a potential method for use in clinical trials.
Collapse
Affiliation(s)
- Sanjar Batirovich Madrakhimov
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research on Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Jin Young Yang
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research on Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Dong Hyuck Ahn
- Laboratory for Translational Research on Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Jung Woo Han
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Tae Ho Ha
- CMLAB, Convergence Technologies for Bio-Medical Science, Seoul, South Korea
| | - Tae Kwann Park
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research on Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Cheonan, Choongchungnam-do, South Korea
- Ex Lumina Therapeutics and Technologies, Bucheon, South Korea
- Corresponding author: Tae Kwann Park, MD, PhD, Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, #170, Jomaru-ro, Wonmi-gu, Bucheon 14584, South Korea.
| |
Collapse
|
20
|
Xu H, Feldman GM, Max EE. High-Dose IV Administration of Rasburicase Suppresses Anti-rasburicase Antibodies, Depletes Rasburicase-Specific Lymphocytes, and Upregulates Treg Cells. AAPS JOURNAL 2020; 22:80. [PMID: 32462555 DOI: 10.1208/s12248-020-00461-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
Therapeutic proteins can be potent agents for treating serious diseases, but in many patients these proteins provoke antibody responses that blunt therapeutic efficacy. Intravenous administration of high doses of some proteins induces immune tolerance, but the mechanisms underlying this effect are poorly understood. As a model to study tolerance induction in mice, we used rasburicase, a commercial recombinant uricase used for the treatment of hyperuricemia. Intraperitoneal (i.p.) injection of rasburicase without or with alum adjuvants induced a clear anti-rasburicase antibody response, but intravenous (i.v.) injection did not. The lack of response to i.v. rasburicase was apparently due to active immune suppression since i.v.-treated mice showed blunted antibody and reduced T cell responses to subsequent i.p. injections of rasburicase. This blunted response was associated with a decrease in rasburicase-specific B cell and T cell responses and an increase in proportion of CD4+ FoxP3+ regulatory T cells (Treg) in the spleen. We examined the number of lymphocytes in peripheral blood after rasburicase i.v. injection. Rasburicase caused a transient reduction in B and T cells, but a robust and sustained depletion of rasburicase-specific B cells. Further experiments showed that rasburicase i.v. injection decreased the number of lymphocytes and was associated with apoptosis of both B cells and activated T cells and that the enhanced percentage of Treg cells was likely mediated by a macrophage-dependent pathway. Thus, our data suggest that apoptosis and depletion of antigen-specific B lymphocytes and upregulation of Treg cells may play important roles in the immune suppression induced by intravenous administration of a therapeutic protein.
Collapse
Affiliation(s)
- Hui Xu
- Laboratory of Immunobiology, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Building 72, Room 2324, 10903 New Hampshire Ave, Silver Spring, Maryland, 20993, USA
| | - Gerald M Feldman
- Laboratory of Immunobiology, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Building 72, Room 2324, 10903 New Hampshire Ave, Silver Spring, Maryland, 20993, USA.
| | - Edward E Max
- Laboratory of Immunobiology, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Building 72, Room 2324, 10903 New Hampshire Ave, Silver Spring, Maryland, 20993, USA
| |
Collapse
|
21
|
Bax BE. Erythrocytes as Carriers of Therapeutic Enzymes. Pharmaceutics 2020; 12:E435. [PMID: 32397259 PMCID: PMC7284836 DOI: 10.3390/pharmaceutics12050435] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/21/2020] [Accepted: 05/06/2020] [Indexed: 02/05/2023] Open
Abstract
Therapeutic enzymes are administered for the treatment of a wide variety of diseases. They exert their effects through binding with a high affinity and specificity to disease-causing substrates to catalyze their conversion to a non-noxious product, to induce an advantageous physiological change. However, the metabolic and clinical efficacies of parenterally or intramuscularly administered therapeutic enzymes are very often limited by short circulatory half-lives and hypersensitive and immunogenic reactions. Over the past five decades, the erythrocyte carrier has been extensively studied as a strategy for overcoming these limitations and increasing therapeutic efficacy. This review examines the rationale for the different therapeutic strategies that have been applied to erythrocyte-mediated enzyme therapy. These strategies include their application as circulating bioreactors, targeting the monocyte-macrophage system, the coupling of enzymes to the surface of the erythrocyte and the engineering of CD34+ hematopoietic precursor cells for the expression of therapeutic enzymes. An overview of the diverse biomedical applications for which they have been investigated is also provided, including the detoxification of exogenous chemicals, thrombolytic therapy, enzyme replacement therapy for metabolic diseases and antitumor therapy.
Collapse
Affiliation(s)
- Bridget E Bax
- Molecular and Clinical Sciences, St. George's, University of London, London SW17 0RE, UK
| |
Collapse
|
22
|
Troy S, Wasilewski M, Beusmans J, Godfrey CJ. Pharmacokinetic Modeling of Intrathecally Administered Recombinant Human Arylsulfatase A (TAK-611) in Children With Metachromatic Leukodystrophy. Clin Pharmacol Ther 2020; 107:1394-1404. [PMID: 31868225 PMCID: PMC7325319 DOI: 10.1002/cpt.1752] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022]
Abstract
Metachromatic leukodystrophy (MLD) is a lysosomal storage disease caused by deficient arylsulfatase A (ASA) activity, which leads to neuronal sulfatide accumulation and motor and cognitive deterioration. Intrathecal delivery of a recombinant human ASA (TAK-611, formerly SHP611) is under development as a potential therapy for MLD. We used serum and cerebrospinal fluid (CSF) TAK-611 concentrations measured during the phase I/II trial of intrathecal TAK-611 to develop a pharmacokinetic (PK) model describing drug disposition. CSF data were well characterized by a two-compartment model in the central nervous system (CNS); a single central compartment described the serum data. Estimated parameters suggested rapid distribution of TAK-611 from CSF into the putative brain tissue compartment, with persistence in the brain between doses (median distributive and terminal half-lives in the CNS: 1.02 and 477 hours, respectively). This model provides a valuable basis for understanding the PK distribution of TAK-611 and for PK/pharmacodynamic analyses of functional outcomes.
Collapse
Affiliation(s)
- Steven Troy
- Shire (a member of the Takeda group of companies), Lexington, Massachusetts, USA.,Radius Health, Waltham, Massachusetts, USA
| | - Margaret Wasilewski
- Shire (a member of the Takeda group of companies), Lexington, Massachusetts, USA.,ID Remedies LLC, Medford, Massachusetts, USA
| | - Jack Beusmans
- Metrum Research Group, Tariffville, Connecticut, USA
| | - C J Godfrey
- Metrum Research Group, Tariffville, Connecticut, USA
| |
Collapse
|
23
|
Mora JR, White JT, DeWall SL. Immunogenicity Risk Assessment for PEGylated Therapeutics. AAPS JOURNAL 2020; 22:35. [PMID: 31993858 DOI: 10.1208/s12248-020-0420-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/07/2020] [Indexed: 12/20/2022]
Abstract
The objective of this manuscript is to provide the reader with two examples on how to present an immunogenicity risk assessment for a PEGylated therapeutic as part of Investigational New Drug (IND) application or during other stages of the drug development process. In order to provide context to the bioanalytical strategies used to support the PEGylated therapeutics presented here, a brief summary of information available for marketed PEGylated biologics is provided. Two case studies are presented, a PEGylated enzyme and a PEGylated growth factor. For the former, the risk assessment covers how to deal with a narrow therapeutic window and suggestions to utilize a PD marker as surrogate for neutralizing antibody assessments in Phase I. The latter has recommendations on additional analytes that should be monitored to mitigate risk of immunogenicity to endogenous counterparts.
Collapse
Affiliation(s)
- Johanna R Mora
- BioAnalytical Sciences, Bristol-Myers Squibb, Princeton, New Jersey, 08543, USA.
| | - Joleen T White
- Drug Metabolism and Pharmacokinetics, EMD Serono, Billerica, Massachusetts, USA
| | - Stephen L DeWall
- Immunogenicity, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| |
Collapse
|
24
|
Nedaeinia R, Faraji H, Javanmard SH, Ferns GA, Ghayour-Mobarhan M, Goli M, Mashkani B, Nedaeinia M, Haghighi MHH, Ranjbar M. Bacterial staphylokinase as a promising third-generation drug in the treatment for vascular occlusion. Mol Biol Rep 2019; 47:819-841. [PMID: 31677034 DOI: 10.1007/s11033-019-05167-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022]
Abstract
Vascular occlusion is one of the major causes of mortality and morbidity. Blood vessel blockage can lead to thrombotic complications such as myocardial infarction, stroke, deep venous thrombosis, peripheral occlusive disease, and pulmonary embolism. Thrombolytic therapy currently aims to rectify this through the administration of recombinant tissue plasminogen activator. Research is underway to design an ideal thrombolytic drug with the lowest risk. Despite the potent clot lysis achievable using approved thrombolytic drugs such as alteplase, reteplase, streptokinase, tenecteplase, and some other fibrinolytic agents, there are some drawbacks, such as high production cost, systemic bleeding, intracranial hemorrhage, vessel re-occlusion by platelet-rich and retracted secondary clots, and non-fibrin specificity. In comparison, bacterial staphylokinase, is a new, small-size plasminogen activator, unlike bacterial streptokinase, it hinders the systemic degradation of fibrinogen and reduces the risk of severe hemorrhage. A fibrin-bound plasmin-staphylokinase complex shows high resistance to a2-antiplasmin-related inhibition. Staphylokinase has the potential to be considered as a promising thrombolytic agent with properties of cost-effective production and the least side effects.
Collapse
Affiliation(s)
- Reza Nedaeinia
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Habibollah Faraji
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran. .,Department of Laboratory Sciences, Faculty of Para-Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Shaghayegh Haghjooye Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Science, Isfahan, Iran
| | - Gordon A Ferns
- Brighton and Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Goli
- Department of Food Science and Technology, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Baratali Mashkani
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mozhdeh Nedaeinia
- Young Researchers and Elite Club, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Mohammad Hossein Hayavi Haghighi
- Department of Health Information Management, Faculty of Para-Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Maryam Ranjbar
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran.,Deputy of Food and Drug, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
25
|
Morlighem JÉRL, Radis-Baptista G. The Place for Enzymes and Biologically Active Peptides from Marine Organisms for Application in Industrial and Pharmaceutical Biotechnology. Curr Protein Pept Sci 2019; 20:334-355. [PMID: 30255754 DOI: 10.2174/1389203719666180926121722] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/10/2018] [Accepted: 09/15/2018] [Indexed: 01/07/2023]
Abstract
Since the beginning of written history, diverse texts have reported the use of enzymatic preparations in food processing and have described the medicinal properties of crude and fractionated venoms to treat various diseases and injuries. With the biochemical characterization of enzymes from distinct sources and bioactive polypeptides from animal venoms, the last sixty years have testified the advent of industrial enzymology and protein therapeutics, which are currently applicable in a wide variety of industrial processes, household products, and pharmaceuticals. Bioprospecting of novel biocatalysts and bioactive peptides is propelled by their unsurpassed properties that are applicable for current and future green industrial processes, biotechnology, and biomedicine. The demand for both novel enzymes with desired characteristics and novel peptides that lead to drug development, has experienced a steady increase in response to the expanding global market for industrial enzymes and peptidebased drugs. Moreover, although largely unexplored, oceans and marine realms, with their unique ecosystems inhabited by a large variety of species, including a considerable number of venomous animals, are recognized as untapped reservoirs of molecules and macromolecules (enzymes and bioactive venom-derived peptides) that can potentially be converted into highly valuable biopharmaceutical products. In this review, we have focused on enzymes and animal venom (poly)peptides that are presently in biotechnological use, and considering the state of prospection of marine resources, on the discovery of useful industrial biocatalysts and drug leads with novel structures exhibiting selectivity and improved performance.
Collapse
Affiliation(s)
- Jean-Étienne R L Morlighem
- Institute for Marine Sciences, Federal University of Ceara, Av da Abolicao 3207. Fortaleza/CE. 60165081, Brazil
| | - Gandhi Radis-Baptista
- Institute for Marine Sciences, Federal University of Ceara, Av da Abolicao 3207. Fortaleza/CE. 60165081, Brazil
| |
Collapse
|
26
|
Production and Purification of Therapeutic Enzymes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1148:1-24. [DOI: 10.1007/978-981-13-7709-9_1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
27
|
Levene M, Pacitti D, Gasson C, Hall J, Sellos-Moura M, Bax BE. Validation of an Immunoassay for Anti-thymidine Phosphorylase Antibodies in Patients with MNGIE Treated with Enzyme Replacement Therapy. Mol Ther Methods Clin Dev 2018; 11:1-8. [PMID: 30294618 PMCID: PMC6170929 DOI: 10.1016/j.omtm.2018.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/21/2018] [Indexed: 02/08/2023]
Abstract
Erythrocyte encapsulated thymidine phosphorylase is recombinant Escherichia coli thymidine phosphorylase encapsulated within human autologous erythrocytes and is under development as an enzyme replacement therapy for the ultra-rare inherited metabolic disorder mitochondrial neurogastrointestinal encephalomyopathy. This study describes the method validation of a two-step bridging electrochemiluminescence immunoassay for the detection of anti-thymidine phosphorylase antibodies in human serum according to current industry practice and regulatory guidelines. The analytical method was assessed for screening cut point, specificity, selectivity, precision, prozone effect, drug tolerance, and stability. Key findings were a correction factor of 129 relative light units for the cut-point determination; a specificity cut point of 93% inhibition; confirmed intra-assay and inter-assay precision; assay sensitivity of 356 ng/mL; no matrix or prozone effects up to 25,900 ng/mL; a drug tolerance of 156 ng/mL; and stability at room temperature for 24 hr and up to five freeze-thaws. Immunogenicity evaluations of serum from three patients who received erythrocyte encapsulated thymidine phosphorylase under a compassionate treatment program showed specific anti-thymidine phosphorylase antibodies in one patient. To conclude, a sensitive, specific, and selective immunoassay has been validated for the measurement of anti-thymidine phosphorylase antibodies; this will be utilized in a phase II pivotal clinical trial of erythrocyte encapsulated thymidine phosphorylase.
Collapse
Affiliation(s)
- Michelle Levene
- Molecular & Clinical Sciences Research Institute, St. George’s, University of London, London, UK
| | - Dario Pacitti
- Molecular & Clinical Sciences Research Institute, St. George’s, University of London, London, UK
| | - Charlotte Gasson
- Biomarker, Bioanalysis and Clinical Sciences, Envigo CRS, Cambridgeshire, UK
| | - Jamie Hall
- Biomarker, Bioanalysis and Clinical Sciences, Envigo CRS, Cambridgeshire, UK
| | | | - Bridget E. Bax
- Molecular & Clinical Sciences Research Institute, St. George’s, University of London, London, UK
- Corresponding author: Bridget E. Bax, Molecular & Clinical Sciences Research Institute, St George’s, University of London, London SW17 0RE, UK.
| |
Collapse
|
28
|
Vu M, Li R, Baskfield A, Lu B, Farkhondeh A, Gorshkov K, Motabar O, Beers J, Chen G, Zou J, Espejo-Mojica AJ, Rodríguez-López A, Alméciga-Díaz CJ, Barrera LA, Jiang X, Ory DS, Marugan JJ, Zheng W. Neural stem cells for disease modeling and evaluation of therapeutics for Tay-Sachs disease. Orphanet J Rare Dis 2018; 13:152. [PMID: 30220252 PMCID: PMC6139903 DOI: 10.1186/s13023-018-0886-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 08/06/2018] [Indexed: 11/30/2022] Open
Abstract
Background Tay-Sachs disease (TSD) is a rare neurodegenerative disorder caused by autosomal recessive mutations in the HEXA gene on chromosome 15 that encodes β-hexosaminidase. Deficiency in HEXA results in accumulation of GM2 ganglioside, a glycosphingolipid, in lysosomes. Currently, there is no effective treatment for TSD. Results We generated induced pluripotent stem cells (iPSCs) from two TSD patient dermal fibroblast lines and further differentiated them into neural stem cells (NSCs). The TSD neural stem cells exhibited a disease phenotype of lysosomal lipid accumulation. The Tay-Sachs disease NSCs were then used to evaluate the therapeutic effects of enzyme replacement therapy (ERT) with recombinant human Hex A protein and two small molecular compounds: hydroxypropyl-β-cyclodextrin (HPβCD) and δ-tocopherol. Using this disease model, we observed reduction of lipid accumulation by employing enzyme replacement therapy as well as by the use of HPβCD and δ-tocopherol. Conclusion Our results demonstrate that the Tay-Sachs disease NSCs possess the characteristic phenotype to serve as a cell-based disease model for study of the disease pathogenesis and evaluation of drug efficacy. The enzyme replacement therapy with recombinant Hex A protein and two small molecules (cyclodextrin and tocopherol) significantly ameliorated lipid accumulation in the Tay-Sachs disease cell model. Electronic supplementary material The online version of this article (10.1186/s13023-018-0886-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mylinh Vu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Rong Li
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Amanda Baskfield
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Billy Lu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Atena Farkhondeh
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Omid Motabar
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Jeanette Beers
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Guokai Chen
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.,Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| | - Jizhong Zou
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Angela J Espejo-Mojica
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Alexander Rodríguez-López
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia.,Chemistry Department, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Carlos J Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Luis A Barrera
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Xuntian Jiang
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Juan J Marugan
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
29
|
Hou ZS, Ulloa-Aguirre A, Tao YX. Pharmacoperone drugs: targeting misfolded proteins causing lysosomal storage-, ion channels-, and G protein-coupled receptors-associated conformational disorders. Expert Rev Clin Pharmacol 2018; 11:611-624. [DOI: 10.1080/17512433.2018.1480367] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Zhi-Shuai Hou
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM) and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| |
Collapse
|
30
|
Lysosomal Targeting Enhancement by Conjugation of Glycopeptides Containing Mannose-6-phosphate Glycans Derived from Glyco-engineered Yeast. Sci Rep 2018; 8:8730. [PMID: 29880804 PMCID: PMC5992200 DOI: 10.1038/s41598-018-26913-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 05/21/2018] [Indexed: 11/08/2022] Open
Abstract
Many therapeutic enzymes for lysosomal storage diseases require a high content of mannose-6-phosphate (M6P) glycan, which is important for cellular uptake and lysosomal targeting. We constructed glyco-engineered yeast harboring a high content of mannosylphosphorylated glycans, which can be converted to M6P glycans by uncapping of the outer mannose residue. In this study, the cell wall of this yeast was employed as a natural M6P glycan source for conjugation to therapeutic enzymes. The extracted cell wall mannoproteins were digested by pronase to generate short glycopeptides, which were further elaborated by uncapping and α(1,2)-mannosidase digestion steps. The resulting glycopeptides containing M6P glycans (M6PgPs) showed proper cellular uptake and lysosome targeting. The purified M6PgPs were successfully conjugated to a recombinant acid α-glucosidase (rGAA), used for the treatment of Pompe disease, by two-step reactions using two hetero-bifunctional crosslinkers. First, rGAA and M6PgPs were modified with crosslinkers containing azide and dibenzocyclooctyne, respectively. In the second reaction using copper-free click chemistry, the azide-functionalized rGAA was conjugated with dibenzocyclooctyne-functionalized M6PgPs without the loss of enzyme activity. The M6PgP-conjugated rGAA had a 16-fold higher content of M6P glycan than rGAA, which resulted in greatly increased cellular uptake and efficient digestion of glycogen accumulated in Pompe disease patient fibroblasts.
Collapse
|
31
|
Kurnaz D, Semerci SY, Babayigit A, Cebeci B, Buyukkale G, Cetinkaya M. Angioedema Associated With Nebulized Recombinant Human DNase in a Preterm Infant: Case Report. J Pediatr Pharmacol Ther 2018; 23:219-222. [DOI: 10.5863/1551-6776-23.3.219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Pulmonary atelectasis leads to difficulties in weaning of the neonates from mechanical ventilation. The management of persistent atelectasis in neonates constitutes a common challenge for physicians. Several reports suggested Recombinant human DNase (rhDNase) as a beneficial therapy for neonates with persistent atelectasis by reducing mucous viscosity. No adverse effect associated with rhDNase treatment was reported in neonates. Herein, we report probable adverse reactions associated with rhDNase use in a preterm infant. Therefore, we suggest that clinicians must be aware of this reaction in neonates and should carefully follow up these infants for the development of adverse reactions. We think that more clinical experience and data are needed to define its tolerability and adverse effect profile in neonates.
Collapse
|
32
|
Abstract
Enzymes are attractive as immunotherapeutics because they can catalyze shifts in the local availability of immunostimulatory and immunosuppressive signals. Clinical success of enzyme immunotherapeutics frequently hinges upon achieving sustained biocatalysis over relevant time scales. The time scale and location of biocatalysis are often dictated by the location of the substrate. For example, therapeutic enzymes that convert substrates distributed systemically are typically designed to have a long half-life in circulation, whereas enzymes that convert substrates localized to a specific tissue or cell population can be more effective when designed to accumulate at the target site. This Topical Review surveys approaches to improve enzyme immunotherapeutic efficacy via chemical modification, encapsulation, and immobilization that increases enzyme accumulation at target sites or extends enzyme half-life in circulation. Examples provided illustrate "replacement therapies" to restore deficient enzyme function, as well as "enhancement therapies" that augment native enzyme function via supraphysiologic doses. Existing FDA-approved enzyme immunotherapies are highlighted, followed by discussion of emerging experimental strategies such as those designed to enhance antitumor immunity or resolve inflammation.
Collapse
Affiliation(s)
- Shaheen A Farhadi
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering , University of Florida , Gainesville , Florida 32611 , United States
| | - Evelyn Bracho-Sanchez
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering , University of Florida , Gainesville , Florida 32611 , United States
| | - Sabrina L Freeman
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering , University of Florida , Gainesville , Florida 32611 , United States
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering , University of Florida , Gainesville , Florida 32611 , United States
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering , University of Florida , Gainesville , Florida 32611 , United States
| |
Collapse
|
33
|
Saeed H, Ali H, Soudan H, Embaby A, El-Sharkawy A, Farag A, Hussein A, Ataya F. Molecular cloning, structural modeling and production of recombinant Aspergillus terreus l. asparaginase in Escherichia coli. Int J Biol Macromol 2018; 106:1041-1051. [DOI: 10.1016/j.ijbiomac.2017.08.110] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/16/2017] [Accepted: 08/17/2017] [Indexed: 11/15/2022]
|
34
|
Scomparin A, Florindo HF, Tiram G, Ferguson EL, Satchi-Fainaro R. Two-step polymer- and liposome-enzyme prodrug therapies for cancer: PDEPT and PELT concepts and future perspectives. Adv Drug Deliv Rev 2017; 118:52-64. [PMID: 28916497 DOI: 10.1016/j.addr.2017.09.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/17/2017] [Accepted: 09/07/2017] [Indexed: 12/14/2022]
Abstract
Polymer-directed enzyme prodrug therapy (PDEPT) and polymer enzyme liposome therapy (PELT) are two-step therapies developed to provide anticancer drugs site-selective intratumoral accumulation and release. Nanomedicines, such as polymer-drug conjugates and liposomal drugs, accumulate in the tumor site due to extravasation-dependent mechanism (enhanced permeability and retention - EPR - effect), and further need to cross the cellular membrane and release their payload in the intracellular compartment. The subsequent administration of a polymer-enzyme conjugate able to accumulate in the tumor tissue and to trigger the extracellular release of the active drug showed promising preclinical results. The development of polymer-enzyme, polymer-drug conjugates and liposomal drugs had undergone a vast advancement over the past decades. Several examples of enzyme mimics for in vivo therapy can be found in the literature. Moreover, polymer therapeutics often present an enzyme-sensitive mechanism of drug release. These nanomedicines can thus be optimal substrates for PDEPT and this review aims to provide new insights and stimuli toward the future perspectives of this promising combination.
Collapse
Affiliation(s)
- Anna Scomparin
- Department of Physiology and Pharmacology, Sackler School of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler School of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel
| | - Elaine L Ferguson
- Advanced Therapies Group, Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler School of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
35
|
Byrne BJ, Geberhiwot T, Barshop BA, Barohn R, Hughes D, Bratkovic D, Desnuelle C, Laforet P, Mengel E, Roberts M, Haroldsen P, Reilley K, Jayaram K, Yang K, Walsh L. A study on the safety and efficacy of reveglucosidase alfa in patients with late-onset Pompe disease. Orphanet J Rare Dis 2017; 12:144. [PMID: 28838325 PMCID: PMC5571484 DOI: 10.1186/s13023-017-0693-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 08/08/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Late-onset Pompe disease is a rare genetic neuromuscular disorder caused by lysosomal acid alpha-glucosidase (GAA) deficiency that ultimately results in mobility loss and respiratory failure. Current enzyme replacement therapy with recombinant human (rh)GAA has demonstrated efficacy in subjects with late-onset Pompe disease. However, long-term effects of rhGAA on pulmonary function have not been observed, likely related to inefficient delivery of rhGAA to skeletal muscle lysosomes and associated deficits in the central nervous system. To address this limitation, reveglucosidase alfa, a novel insulin-like growth factor 2 (IGF2)-tagged GAA analogue with improved lysosomal uptake, was developed. This study evaluated the pharmacokinetics, safety, and exploratory efficacy of reveglucosidase alfa in 22 subjects with late-onset Pompe disease who were previously untreated with rhGAA. RESULTS Reveglucosidase alfa plasma concentrations increased linearly with dose, and the elimination half-life was <1.2 h. Eighteen of 22 subjects completed 72 weeks of treatment. The most common adverse events were hypoglycemia (63%), dizziness, fall, headache, and nausea (55% for each). Serious adverse events included hypersensitivity (n = 1), symptomatic hypoglycemia (n = 2), presyncope (n = 1), and acute cardiac failure (n = 1). In the dose-escalation study, all treated subjects tested positive for anti-reveglucosidase alfa, anti-rhGAA, anti-IGF1, and anti-IGF2 antibodies at least once. Subjects receiving 20 mg/kg of reveglucosidase alfa demonstrated increases in predicted maximum inspiratory pressure (13.9%), predicted maximum expiratory pressure (8.0%), forced vital capacity (-0.4%), maximum voluntary ventilation (7.4 L/min), and mean absolute walking distance (22.3 m on the 6-min walk test) at 72 weeks. CONCLUSIONS Additional studies are needed to further assess the safety and efficacy of this approach. Improvements in respiratory muscle strength, lung function, and walking endurance in subjects with LOPD may make up for the risk of hypersensitivity reactions and hypoglycemia. Reveglucosidase alfa may provide a new treatment option for patients with late-onset Pompe disease. TRIAL REGISTRATION ISRCTN01435772 and ISRCTN01230801 , registered 27 October 2011.
Collapse
Affiliation(s)
- Barry J Byrne
- University of Florida, School of Medicine, 1600 SW Archer Road, Gainesville, FL, 32607, USA. .,Department of Pediatrics, University of Florida, P.O. Box 100296, Gainesville, FL, 32610, USA.
| | - Tarekegn Geberhiwot
- University Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2GW, UK
| | - Bruce A Barshop
- University of California San Diego Health System, 4168 Front Street, San Diego, CA, 92103, USA
| | - Richard Barohn
- Kansas University Medical Center, 3901 Rainbow Blvd/MSN 2012, Kansas City, KS, 66160, USA
| | - Derralynn Hughes
- Royal Free London NHS Foundation & University College London Department of Hematology, Pond St, London, NW3 2QG, UK
| | | | | | - Pascal Laforet
- Paris-Est Neuromuscular Center, INSERM U974, UPMC, Hôpital Pitié-Salpêtrière, 47-83 boulevard de l'Hôpital, 75013, Paris, France
| | - Eugen Mengel
- Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Mark Roberts
- Salford Royal NHS Foundation Trust, M6 8HD, Salford, UK
| | - Peter Haroldsen
- BioMarin Pharmaceutical, 105 Digital Drive, Novato, CA, 94949, USA
| | - Kristin Reilley
- BioMarin Pharmaceutical, 105 Digital Drive, Novato, CA, 94949, USA
| | - Kala Jayaram
- BioMarin Pharmaceutical, 105 Digital Drive, Novato, CA, 94949, USA
| | - Ke Yang
- BioMarin Pharmaceutical, 105 Digital Drive, Novato, CA, 94949, USA
| | - Liron Walsh
- BioMarin Pharmaceutical, 105 Digital Drive, Novato, CA, 94949, USA
| | | |
Collapse
|
36
|
Majtan T, Park I, Carrillo RS, Bublil EM, Kraus JP. Engineering and Characterization of an Enzyme Replacement Therapy for Classical Homocystinuria. Biomacromolecules 2017; 18:1747-1761. [DOI: 10.1021/acs.biomac.7b00154] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Tomas Majtan
- Department
of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Insun Park
- Department
of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Richard S. Carrillo
- Department
of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Erez M. Bublil
- Orphan
Technologies Ltd., Rapperswil, CH-8640, Switzerland
| | - Jan P. Kraus
- Department
of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| |
Collapse
|
37
|
Donida B, Jacques CED, Mescka CP, Rodrigues DGB, Marchetti DP, Ribas G, Giugliani R, Vargas CR. Oxidative damage and redox in Lysosomal Storage Disorders: Biochemical markers. Clin Chim Acta 2017; 466:46-53. [DOI: 10.1016/j.cca.2017.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 01/04/2017] [Accepted: 01/07/2017] [Indexed: 02/03/2023]
|
38
|
Keeling KM. Nonsense Suppression as an Approach to Treat Lysosomal Storage Diseases. Diseases 2016; 4:32. [PMID: 28367323 PMCID: PMC5370586 DOI: 10.3390/diseases4040032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/14/2016] [Indexed: 02/08/2023] Open
Abstract
In-frame premature termination codons (PTCs) (also referred to as nonsense mutations) comprise ~10% of all disease-associated gene lesions. PTCs reduce gene expression in two ways. First, PTCs prematurely terminate translation of an mRNA, leading to the production of a truncated polypeptide that often lacks normal function and/or is unstable. Second, PTCs trigger degradation of an mRNA by activating nonsense-mediated mRNA decay (NMD), a cellular pathway that recognizes and degrades mRNAs containing a PTC. Thus, translation termination and NMD are putative therapeutic targets for the development of treatments for genetic diseases caused by PTCs. Over the past decade, significant progress has been made in the identification of compounds with the ability to suppress translation termination of PTCs (also referred to as readthrough). More recently, NMD inhibitors have also been explored as a way to enhance the efficiency of PTC suppression. Due to their relatively low threshold for correction, lysosomal storage diseases are a particularly relevant group of diseases to investigate the feasibility of nonsense suppression as a therapeutic approach. In this review, the current status of PTC suppression and NMD inhibition as potential treatments for lysosomal storage diseases will be discussed.
Collapse
Affiliation(s)
- Kim M Keeling
- Department of Biochemistry and Molecular Genetics, Gregory Fleming Cystic Fibrosis Research Center, Comprehensive Arthritis, Musculoskeletal, Bone, and Autoimmunity Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA; ; Tel.: +1-205-975-6585
| |
Collapse
|
39
|
Won JS, Singh AK, Singh I. Biochemical, cell biological, pathological, and therapeutic aspects of Krabbe's disease. J Neurosci Res 2016; 94:990-1006. [PMID: 27638584 PMCID: PMC5812347 DOI: 10.1002/jnr.23873] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/01/2016] [Accepted: 07/14/2016] [Indexed: 12/14/2022]
Abstract
Krabbe's disease (KD; also called globoid cell leukodystrophy) is a genetic disorder involving demyelination of the central (CNS) and peripheral (PNS) nervous systems. The disease may be subdivided into three types, an infantile form, which is the most common and severe; a juvenile form; and a rare adult form. KD is an autosomal recessive disorder caused by a deficiency of galactocerebrosidase activity in lysosomes, leading to accumulation of galactoceramide and neurotoxic galactosylsphingosine (psychosine [PSY]) in macrophages (globoid cells) as well as neural cells, especially in oligodendrocytes and Schwann cells. This ultimately results in damage to myelin in both CNS and PNS with associated morbidity and mortality. Accumulation of PSY, a lysolipid with detergent-like properties, over a threshold level could trigger membrane destabilization, leading to cell lysis. Moreover, subthreshold concentrations of PSY trigger cell signaling pathways that induce oxidative stress, mitochondrial dysfunction, apoptosis, inflammation, endothelial/vascular dysfunctions, and neuronal and axonal damage. From the time the "psychosine hypothesis" was proposed, considerable efforts have been made in search of an effective therapy for lowering PSY load with pharmacological, gene, and stem cell approaches to attenuate PSY-induced neurotoxicity. This Review focuses on the recent advances and prospective research for understanding disease mechanisms and therapeutic approaches for KD. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Je-Seong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Avtar K. Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
- Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
40
|
Bonaventura A, Montecucco F, Dallegri F. Update on the effects of treatment with recombinant tissue-type plasminogen activator (rt-PA) in acute ischemic stroke. Expert Opin Biol Ther 2016; 16:1323-1340. [PMID: 27548625 DOI: 10.1080/14712598.2016.1227779] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Acute ischemic stroke (AIS) represents a major cause of death and disability all over the world. The recommended therapy aims at dissolving the clot to re-establish quickly the blood flow to the brain and reduce neuronal injury. Intravenous administration of recombinant tissue-type plasminogen activator (rt-PA) is clinically used with this goal. AREAS COVERED A description of beneficial and detrimental effects of rt-PA treatment is addressed. An overview of new therapies against AIS, such as new thrombolytics, sonolysis and sonothrombolysis, endovascular procedures, and association therapies is provided. Updates on the pathophysiological process leading to intracranial hemorrhage (ICH) is also discussed. EXPERT OPINION rt-PA treatment in AIS patients is beneficial to recovery outcomes. To weaken risks and improve benefits, it might be relevant to consider: i) a definitive identification of risk factors for symptomatic ICH; ii). a better organization of the health care system to reduce time-to-treatment and enhance discharge management. The pharmacological improvement of new thrombolytic drugs (such as tenecteplase and desmoteplase) targeting harmful and maximally exploiting beneficial effects might further reduce mortality and disability in AIS.
Collapse
Affiliation(s)
- Aldo Bonaventura
- a First Clinic of Internal Medicine, Department of Internal Medicine , University of Genoa School of Medicine , Genoa , Italy.,b IRCCS AOU San Martino - IST, Genoa , Genoa , Italy
| | - Fabrizio Montecucco
- a First Clinic of Internal Medicine, Department of Internal Medicine , University of Genoa School of Medicine , Genoa , Italy.,b IRCCS AOU San Martino - IST, Genoa , Genoa , Italy.,c Centre of Excellence for Biomedical Research (CEBR) , University of Genoa , Genoa , Italy
| | - Franco Dallegri
- a First Clinic of Internal Medicine, Department of Internal Medicine , University of Genoa School of Medicine , Genoa , Italy.,b IRCCS AOU San Martino - IST, Genoa , Genoa , Italy
| |
Collapse
|
41
|
Xiong H, Zhou Y, Zhou Q, He D, Deng X, Sun Q, Zhang J. Nanocapsule assemblies as effective enzyme delivery systems against hyperuricemia. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1557-66. [DOI: 10.1016/j.nano.2016.02.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 02/03/2016] [Accepted: 02/08/2016] [Indexed: 12/14/2022]
|
42
|
Oh DB. Glyco-engineering strategies for the development of therapeutic enzymes with improved efficacy for the treatment of lysosomal storage diseases. BMB Rep 2016; 48:438-44. [PMID: 25999178 PMCID: PMC4576951 DOI: 10.5483/bmbrep.2015.48.8.101] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Indexed: 11/20/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of inherent diseases characterized by massive accumulation of undigested compounds in lysosomes, which is caused by genetic defects resulting in the deficiency of a lysosomal hydrolase. Currently, enzyme replacement therapy has been successfully used for treatment of 7 LSDs with 10 approved therapeutic enzymes whereas new approaches such as pharmacological chaperones and gene therapy still await evaluation in clinical trials. While therapeutic enzymes for Gaucher disease have N-glycans with terminal mannose residues for targeting to macrophages, the others require N-glycans containing mannose-6-phosphates that are recognized by mannose-6-phosphate receptors on the plasma membrane for cellular uptake and targeting to lysosomes. Due to the fact that efficient lysosomal delivery of therapeutic enzymes is essential for the clearance of accumulated compounds, the suitable glycan structure and its high content are key factors for efficient therapeutic efficacy. Therefore, glycan remodeling strategies to improve lysosomal targeting and tissue distribution have been highlighted. This review describes the glycan structures that are important for lysosomal targeting and provides information on recent glyco-engineering technologies for the development of therapeutic enzymes with improved efficacy. [BMB Reports 2015; 48(8): 438-444]
Collapse
Affiliation(s)
- Doo-Byoung Oh
- Synthetic Biology and Bioengineering Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB); Biosystems and Bioengineering Program, University of Science and Technology (UST), Daejeon 34141, Korea
| |
Collapse
|
43
|
Schuchman EH. Acid ceramidase and the treatment of ceramide diseases: The expanding role of enzyme replacement therapy. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1459-71. [PMID: 27155573 DOI: 10.1016/j.bbadis.2016.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/18/2016] [Accepted: 05/03/2016] [Indexed: 01/20/2023]
Abstract
Ceramides are a diverse group of sphingolipids that play important roles in many biological processes. Acid ceramidase (AC) is one key enzyme that regulates ceramide metabolism. Early research on AC focused on the fact that it is the enzyme deficient in the rare genetic disorder, Farber Lipogranulomatosis. Recent research has revealed that deficiency of the same enzyme is responsible for a rare form of spinal muscular atrophy associated with myoclonic epilepsy (SMA-PME). Due to their diverse role in biology, accumulation of ceramides also has been implicated in the pathobiology of many other common diseases, including infectious lung diseases, diabetes, cancers and others. This has revealed the potential of AC as a therapy for many of these diseases. This review will focus on the biology of AC and the potential role of this enzyme in the treatment of human disease.
Collapse
Affiliation(s)
- Edward H Schuchman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
44
|
Aronson JK. Defining 'nutraceuticals': neither nutritious nor pharmaceutical. Br J Clin Pharmacol 2016; 83:8-19. [PMID: 26991455 DOI: 10.1111/bcp.12935] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 03/09/2016] [Indexed: 12/19/2022] Open
Abstract
There are widespread inconsistencies and contradictions in the many published definitions of 'nutraceuticals' and 'functional foods', demonstrating wholesale uncertainty about what they actually are. Furthermore, in a 2014 lecture, the inventor of the term 'nutraceutical', confessing that nutraceuticals do not work, said that 'the quest to demonstrate whether … long-term supplementation [with nutraceuticals] can prevent serious diseases … has come to an end'. Definitions of 'nutraceuticals' and related terms, still widely used, should therefore be explored systematically. There are no internationally agreed definitions of 'nutraceuticals' and 'functional foods', or of similar terms, such as 'health foods', or of terms related to herbal products, which are sometimes referred to as 'nutraceuticals', compounding the confusion. 'Nutraceuticals' and 'functional foods' are vague, nondiscriminatory, unhelpful terms; the evidence suggests that they should be abandoned in favour of more precise terms. The term 'dietary supplement' is widely used to designate formulations that are also called 'nutraceuticals' but it would be better restricted to individual compounds used to treat or prevent deficiencies. 'Fortified foods', sometimes called 'designer foods', are foods to which compounds of proven therapeutic or preventive efficacy (e.g. folic acid) have been added. Other terms, such as 'food', 'foodstuffs', 'eat', 'drink', and 'nutrition', are well defined, as are 'medicinal products' and 'pharmaceutical formulations'. Dietary regimens, such as Mediterranean or nitrate-rich diets or vegetarianism, can affect health. A dietary regimen of this kind can be defined as a programme of food, of a defined kind and/or quantity, prescribed or adopted for the restoration or preservation of health.
Collapse
Affiliation(s)
- Jeffrey K Aronson
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, Oxford, OX26GG, UK
| |
Collapse
|
45
|
Fang Y, Fullwood MJ. Roles, Functions, and Mechanisms of Long Non-coding RNAs in Cancer. GENOMICS PROTEOMICS & BIOINFORMATICS 2016; 14:42-54. [PMID: 26883671 PMCID: PMC4792843 DOI: 10.1016/j.gpb.2015.09.006] [Citation(s) in RCA: 739] [Impact Index Per Article: 92.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 08/31/2015] [Accepted: 09/17/2015] [Indexed: 12/28/2022]
Abstract
Long non-coding RNAs (lncRNAs) play important roles in cancer. They are involved in chromatin remodeling, as well as transcriptional and post-transcriptional regulation, through a variety of chromatin-based mechanisms and via cross-talk with other RNA species. lncRNAs can function as decoys, scaffolds, and enhancer RNAs. This review summarizes the characteristics of lncRNAs, including their roles, functions, and working mechanisms, describes methods for identifying and annotating lncRNAs, and discusses future opportunities for lncRNA-based therapies using antisense oligonucleotides.
Collapse
Affiliation(s)
- Yiwen Fang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Melissa J Fullwood
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; Yale-NUS Liberal Arts College, Singapore 138527, Singapore.
| |
Collapse
|
46
|
Degreef I. Collagenase Treatment in Dupuytren Contractures: A Review of the Current State Versus Future Needs. Rheumatol Ther 2016; 3:43-51. [PMID: 27747514 DOI: 10.1007/s40744-016-0027-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Indexed: 11/26/2022] Open
Abstract
Dupuytren disease is highly prevalent and the finger contractures can be very extensile, compromising the patients' hand function. To restore full function, contractures have been addressed by cutting the causative strands for nearly 200 years, ever since Baron Guillaume Dupuytren demonstrated his technique at the beginning of the nineteenth century. Surgery can be minimal (fasciotomy) or quite invasive (fasciectomy and even skin replacement). However, in the last decade translational research has introduced the non-surgical technique of enzymatic fasciotomy with collagenase injections. Now, finger contractures can be released with single injections on monthly intervals, to address one joint contracture at a time. However, in hands affected with Dupuytren contractures to the extent that the patient calls for treatment, most often more than one joint is involved. In surgical treatment options all contracted joints are addressed in a single procedure. Nevertheless, extensile surgery withholds inherent risks of complications and intense rehabilitation. Today, the minimally-invasive method with enzymatic fasciotomy by collagenase injection has demonstrated reliable outcomes with few morbidities and early recovery. However, single-site injection is todays' standard procedure and multiple joints are addressed in several sessions with monthly intervals. This triggers a longer recovery and treatment burden in severely affected hands even though surgery is avoided. Therefore, further treatment modalities of collagenase use are explored. Adjustments in the treatment regimes' flexibility and collagenase injections addressing more than one joint contracture simultaneously will improve the burden of multiple sessions and, therefore, enzymatic fasciotomy may become the preferred method in more extensile Dupuytren contractures. In this independent review, the challenge of Dupuytren disease affecting a single versus multiple joints is presented. The pros and cons of collagenase use are weighed, founded by the available scientific background. The demands and options for collagenase in future treatment regimens for extensile Dupuytren contractures are discussed.
Collapse
Affiliation(s)
- Ilse Degreef
- Orthopedic Department, Hand Surgery, University Hospitals Leuven, Louvain, Belgium.
| |
Collapse
|