1
|
Jülke EM, Beck-Sickinger AG. Peptide therapeutics: current status and future opportunity with focus on nose-to-brain delivery☆. Peptides 2025; 188:171404. [PMID: 40222598 DOI: 10.1016/j.peptides.2025.171404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Peptide drugs are a highly diverse group of therapeutic agents. Over the last decade, more than 40 peptides have been approved for clinical use. They target different structures, ranging from G protein-coupled receptors (GPCRs) to pathogens and are used to treat a variety of indications, including metabolic disorders, genetic diseases, acute illnesses and more. Structurally, peptide therapeutics are a heterogeneous class. This diversity allows them to bridge the gap between small molecules and biologics. However, limited metabolic stability and bioavailability must be addressed. Strategies to improve the half-life include backbone and sequence modification, cyclization and the addition of stabilizing moieties. Great strides have been made in recent years towards achieving sufficient drug uptake for oral application have been achieved within recent years. However, these methods require specialized peptide design or involve permeabilization of the gastrointestinal tract. Consequently, other routes of administration are being explored. One promising approach is the nasal application of peptides. This method can be used for systemic uptake, but also allows for direct nose-to-brain delivery of compounds. While successful nose-to-brain delivery is already used in the clinic, the underlining mechanisms are poorly understood. Strategies for rational optimization are needed to make this method more applicable to a wider range of compounds. Overall, approved peptide therapeutics cover a wide range of applications and have demonstrated a growing and novel potential in recent drug discovery.
Collapse
Affiliation(s)
- Eva-Maria Jülke
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, Leipzig 04103, Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, Leipzig 04103, Germany.
| |
Collapse
|
2
|
Ago Y, Khan S, Klipner K, Bradford A, Tomatsu S. Identification of Surrogate Biomarkers for Mucopolysaccharidosis Type IVA. Int J Mol Sci 2025; 26:4940. [PMID: 40430081 PMCID: PMC12112068 DOI: 10.3390/ijms26104940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/12/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Mucopolysaccharidosis type IVA (MPS IVA, Morquio A syndrome) is a rare inherited disorder characterized by skeletal dysplasia due to deficient N-acetylgalactosamine-6-sulfate sulfatase activity, resulting in glycosaminoglycan (GAG) accumulation. Identifying accurate biomarkers reflecting clinical severity and therapeutic response remains challenging. This study evaluated potential surrogate biomarkers, including N-terminal pro-C-type natriuretic peptide (NT-proCNP), collagen types I and II, mono-sulfated keratan sulfate (KS), di-sulfated KS, and chondroitin-6-sulfate (C6S), in blood and urine samples from 60 patients ranging from 1 to 62 years of age. NT-proCNP levels were significantly elevated in patients of all ages and negatively correlated with growth impairment, especially after 8 years of age. Collagen type I levels significantly increased in adult patients, whereas collagen type II showed age-dependent elevations. Urinary KS, in mono- and di-sulfated forms, demonstrated moderate negative correlations with growth impairment. Moreover, NT-proCNP, mono- and di-sulfated KS in plasma, and urinary di-sulfated KS were not affected by enzyme replacement therapy in patients younger than 12 years, unlike urinary mono-sulfated KS. In conclusion, NT-proCNP has emerged as a promising independent biomarker reflecting the severity of skeletal dysplasia and possibly the near-future growth rate. These findings highlight the potential role of NT-proCNP in clinical assessment and monitoring therapeutic efficacy, addressing current unmet needs in MPS IVA management.
Collapse
Affiliation(s)
- Yasuhiko Ago
- Nemours Children’s Health, Wilmington, DE 19803, USA; (Y.A.); (S.K.); (K.K.); (A.B.)
| | - Shaukat Khan
- Nemours Children’s Health, Wilmington, DE 19803, USA; (Y.A.); (S.K.); (K.K.); (A.B.)
| | - Kimberly Klipner
- Nemours Children’s Health, Wilmington, DE 19803, USA; (Y.A.); (S.K.); (K.K.); (A.B.)
| | - Allison Bradford
- Nemours Children’s Health, Wilmington, DE 19803, USA; (Y.A.); (S.K.); (K.K.); (A.B.)
| | - Shunji Tomatsu
- Nemours Children’s Health, Wilmington, DE 19803, USA; (Y.A.); (S.K.); (K.K.); (A.B.)
- Faculty of Arts and Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu 501-1193, Japan
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
3
|
Kim T, Baek E, Kim J. Exploring Macrocyclic Chemical Space: Strategies and Technologies for Drug Discovery. Pharmaceuticals (Basel) 2025; 18:617. [PMID: 40430438 PMCID: PMC12114740 DOI: 10.3390/ph18050617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Macrocycles have emerged as significant therapeutic candidates in drug discovery due to their unique capacity to target complex and traditionally inaccessible biological interfaces. Their structurally constrained three-dimensional configurations facilitate high-affinity interactions with challenging targets, notably protein-protein interfaces. However, despite their potential, the synthesis and optimization of macrocyclic compounds present considerable challenges related to structural complexity, synthetic accessibility, and the attainment of favorable drug-like properties, particularly cell permeability and oral bioavailability. Recent advancements in synthetic methodologies have expanded the chemical space accessible to macrocycles, enabling the creation of structurally diverse and pharmacologically active compounds. Concurrent developments in computational strategies have further enhanced macrocycle design, providing valuable insights into structural optimization and predicting molecular properties essential for therapeutic efficacy. Additionally, a deeper understanding of macrocycles' conformational adaptability, especially their ability to internally shield polar functionalities to improve membrane permeability, has significantly informed their rational design. This review discusses recent innovations in synthetic and computational methodologies that have advanced macrocycle drug discovery over the past five years. It emphasizes the importance of integrating these strategies to overcome existing challenges, illustrating how their synergy expands the therapeutic potential and chemical diversity of macrocycles. Selected case studies underscore the practical impact of these integrated approaches, highlighting promising therapeutic applications across diverse biomedical targets.
Collapse
Affiliation(s)
- Taegwan Kim
- Department of Chemistry, Integrative Institute of Basic Science, Soongsil University, Seoul 06978, Republic of Korea;
| | - Eunbee Baek
- Department of Green Chemistry and Materials Engineering, Soongsil University, Seoul 06978, Republic of Korea;
| | - Jonghoon Kim
- Department of Chemistry, Integrative Institute of Basic Science, Soongsil University, Seoul 06978, Republic of Korea;
- Department of Green Chemistry and Materials Engineering, Soongsil University, Seoul 06978, Republic of Korea;
| |
Collapse
|
4
|
Sadamori K, Kubo T, Yoshida T, Yamamoto M, Shibata Y, Fukasawa K, Tokumura K, Horie T, Kadota T, Yamakawa R, Hojo H, Tanaka N, Kitao T, Shirahase H, Hinoi E. CDK8 inhibitor KY-065 rescues skeletal abnormalities in achondroplasia model mice. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167626. [PMID: 39674288 DOI: 10.1016/j.bbadis.2024.167626] [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: 06/01/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024]
Abstract
Cyclin-dependent kinase 8 (CDK8) is a transcription-related CDK family member implicated in the regulation of bone homeostasis, and we recently demonstrated that our internally developed CDK8 inhibitor KY-065 can prevent postmenopausal osteoporosis in a mouse model. Achondroplasia (ACH), the most common form of genetic dwarfism in humans, is caused by a gain-of-function mutation in fibroblast growth factor receptor 3 (FGFR3), a receptor tyrosine kinase that activates downstream mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription (STAT) signaling pathways. The first precision drug approved for the treatment of ACH in children, the C-type natriuretic peptide analog vosoritide, antagonizes the MAPK pathway, while there are currently no effective and safe medications targeting the STAT1 pathway. Here, we demonstrate that KY-065 rescues impaired chondrogenesis and stunted long bone growth in the Fgfr3Ach mouse model of ACH. KY-065 inhibited CDK8 with high affinity in vitro by competing with ATP. The CDK8 expression and STAT1Ser727 phosphorylation were upregulated in chondrocytes isolated from ACH model mice, and KY-065 repressed its phosphorylation and restored normal chondrogenic differentiation without affecting MAPK activation. Moreover, daily administration of 10 mg/kg KY-065 to Fgfr3Ach mice (yielding a peak concentration of 22.0 ± 1.47 μM in plasma) resulted in significant elongation of long bone and improved growth plate cytoarchitecture. Collectively, these findings identify the CDK8 in chondrocytes as a potential therapeutic target for ACH and KY-065 as a promising candidate drug treatment for this debilitating skeletal disease.
Collapse
Affiliation(s)
- Koki Sadamori
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Takuya Kubo
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tomoki Yoshida
- School of Pharmacy, Kitasato University, Tokyo 108-8641, Japan
| | - Megumi Yamamoto
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Ltd., Kyoto, Japan
| | - Yui Shibata
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Ltd., Kyoto, Japan
| | - Kazuya Fukasawa
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Kazuya Tokumura
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tetsuhiro Horie
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Takuya Kadota
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Ryotaro Yamakawa
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Hironori Hojo
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Nobutada Tanaka
- School of Pharmacy, Kitasato University, Tokyo 108-8641, Japan
| | - Tatsuya Kitao
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Ltd., Kyoto, Japan
| | - Hiroaki Shirahase
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Ltd., Kyoto, Japan
| | - Eiichi Hinoi
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1196, Japan; Center for One Medicine Innovative Translational Research (COMIT), Division of Innovative Modality Development, Gifu University, Gifu 501-1196, Japan.
| |
Collapse
|
5
|
Martian PC, Tertis M, Leonte D, Hadade N, Cristea C, Crisan O. Cyclic peptides: A powerful instrument for advancing biomedical nanotechnologies and drug development. J Pharm Biomed Anal 2025; 252:116488. [PMID: 39388867 DOI: 10.1016/j.jpba.2024.116488] [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: 07/18/2024] [Revised: 09/05/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
Cyclic peptides have emerged as an essential tool in the advancement of biomedical nanotechnologies, offering unique structural and functional advantages over linear peptides. This review article aims to highlight the roles of cyclic peptides in the development of biomedical fields, with a particular focus on their application in drug discovery and delivery. Cyclic peptides exhibit exceptional stability, bioavailability, and binding specificity, making them ideal candidates for therapeutic and diagnostic applications. We explore the synthesis and design strategies that enable the precise control of cyclic peptide structures, leading to enhanced performance in targeting specific cellular pathways. The article also highlights recent breakthroughs in the use of cyclic peptides for creating innovative drug delivery systems, including nanoparticle conjugates and peptide-drug conjugates, which have shown promise in improving the efficacy and safety profiles of existing traditional treatments. The integration of cyclic peptides into nanotechnological frameworks holds significant promise for addressing unmet medical needs, providing a foundation for future advancements in personalized medicine and targeted drug delivery.
Collapse
Affiliation(s)
- Paul Cristian Martian
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 4 Pasteur Street, Cluj-Napoca 400021, Romania
| | - Mihaela Tertis
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 4 Pasteur Street, Cluj-Napoca 400021, Romania
| | - Denisa Leonte
- Department of Organic Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 28 Victor Babes Street, Cluj-Napoca 400023, Romania
| | - Niculina Hadade
- Department of Chemistry, Faculty of Chemistry and Chemical Engineering, Babes Bolyai University, 11 Arany Janos Street, Cluj-Napoca 400028, Romania
| | - Cecilia Cristea
- Department of Analytical Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 4 Pasteur Street, Cluj-Napoca 400021, Romania.
| | - Ovidiu Crisan
- Department of Organic Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 28 Victor Babes Street, Cluj-Napoca 400023, Romania
| |
Collapse
|
6
|
Sato Y, Kawamura K. C-type natriuretic peptide promotes human granulosa cell growth and estradiol production: Implications for early follicle development. Reprod Med Biol 2025; 24:e12626. [PMID: 39845482 PMCID: PMC11751860 DOI: 10.1002/rmb2.12626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/21/2024] [Indexed: 01/24/2025] Open
Abstract
Purpose To investigate the effects of C-type natriuretic peptide (CNP) on human granulosa cell growth and elucidate its regulatory mechanisms. Methods A human non-luteinizing granulosa cell line (HGrC) developed from small antral follicles was used to assess the impact of CNP on cell proliferation and estrogen synthesis. cGMP production via the guanylate cyclase domain of the CNP receptor, natriuretic peptide receptor 2 (NPR2), was confirmed. The regulation of CNP encoding natriuretic peptide C (NPPC) and NPR2 by estradiol and oocyte-derived factors (ODFs) was examined. Results Besides detecting both NPPC and NPR2, CNP increased cellular proliferation. The specific action of CNP on cell proliferation was confirmed using siRNA transfection. CNP stimulated cGMP production, whereas a guanylate-cyclase inhibitor suppressed CNP-induced cell proliferation. Estradiol production was elevated by CNP treatment, accompanied by increased expression of estrogen synthetic enzymes. Furthermore, CNP upregulated NPR2 expression in cooperation with estradiol and ODFs, while estradiol increased NPPC expression. Conclusion This study demonstrates CNP stimulation of human granulosa cell growth and suggests potential cross-talk between these cells and oocytes. Further research on the simultaneous administration of CNP and estradiol may offer a promising approach for promoting early-stage follicle development in infertility treatments for patients with poor ovarian reserve.
Collapse
Affiliation(s)
- Yorino Sato
- Department of Obstetrics and GynecologyJuntendo University Faculty of MedicineBunkyoTokyoJapan
| | - Kazuhiro Kawamura
- Department of Obstetrics and GynecologyJuntendo University Faculty of MedicineBunkyoTokyoJapan
| |
Collapse
|
7
|
Dickinson YA, Moyes AJ, Hobbs AJ. C-type natriuretic peptide (CNP): The cardiovascular system and beyond. Pharmacol Ther 2024; 262:108708. [PMID: 39154787 DOI: 10.1016/j.pharmthera.2024.108708] [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: 03/28/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
C-type natriuretic peptide (CNP) represents the 'local' member of the natriuretic peptide family, functioning in an autocrine or paracrine capacity to modulate a hugely diverse portfolio of physiological processes. Whilst the best-characterised of these regulatory roles are in the cardiovascular system, akin to its predominantly endocrine siblings atrial (ANP) and brain (BNP) natriuretic peptides, CNP governs many additional, unrelated mechanisms including bone growth, gamete maturation, auditory processing, and neuronal integrity. Furthermore, there is currently great interest in mimicking the biological activity of CNP for therapeutic gain in many of these disparate organ systems. Herein, we provide an overview of the physiology, pathophysiology and pharmacology of CNP in both cardiovascular and non-cardiovascular settings.
Collapse
Affiliation(s)
- Yasmin A Dickinson
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Amie J Moyes
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Adrian J Hobbs
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
8
|
Lu Z, Verginadis I, Kumazoe M, Castillo GM, Yao Y, Guerra RE, Bicher S, You M, McClung G, Qiu R, Xiao Z, Miao Z, George SS, Beiting DP, Nojiri T, Tanaka Y, Fujimura Y, Onda H, Hatakeyama Y, Nishimoto-Ashfield A, Bykova K, Guo W, Fan Y, Buynov NM, Diehl JA, Stanger BZ, Tachibana H, Gade TP, Puré E, Koumenis C, Bolotin EM, Fuchs SY. Modified C-type natriuretic peptide normalizes tumor vasculature, reinvigorates antitumor immunity, and improves solid tumor therapies. Sci Transl Med 2024; 16:eadn0904. [PMID: 39167664 PMCID: PMC11866103 DOI: 10.1126/scitranslmed.adn0904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/23/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024]
Abstract
Deficit of oxygen and nutrients in the tumor microenvironment (TME) triggers abnormal angiogenesis that produces dysfunctional and leaky blood vessels, which fail to adequately perfuse tumor tissues. Resulting hypoxia, exacerbation of metabolic disturbances, and generation of an immunosuppressive TME undermine the efficacy of anticancer therapies. Use of carefully scheduled angiogenesis inhibitors has been suggested to overcome these problems and normalize the TME. Here, we propose an alternative agonist-based normalization approach using a derivative of the C-type natriuretic peptide (dCNP). Multiple gene expression signatures in tumor tissues were affected in mice treated with dCNP. In several mouse orthotopic and subcutaneous solid tumor models including colon and pancreatic adenocarcinomas, this well-tolerated agent stimulated formation of highly functional tumor blood vessels to reduce hypoxia. Administration of dCNP also inhibited stromagenesis and remodeling of the extracellular matrix and decreased tumor interstitial fluid pressure. In addition, treatment with dCNP reinvigorated the antitumor immune responses. Administration of dCNP decelerated growth of primary mouse tumors and suppressed their metastases. Moreover, inclusion of dCNP into the chemo-, radio-, or immune-therapeutic regimens increased their efficacy against solid tumors in immunocompetent mice. These results demonstrate the proof of principle for using vasculature normalizing agonists to improve therapies against solid tumors and characterize dCNP as the first in class among such agents.
Collapse
Affiliation(s)
- Zhen Lu
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ioannis Verginadis
- Dept. of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Motofumi Kumazoe
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | | | - Yao Yao
- PharmaIN Corp., Bothell, WA 98011, USA
| | | | - Sandra Bicher
- Dept. of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Menghao You
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - George McClung
- Dept. of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rong Qiu
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zebin Xiao
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhen Miao
- Dept. of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Subin S. George
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P. Beiting
- Dept. of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Takashi Nojiri
- Dept. of General Thoracic Surgery, Higashiosaka City Medical Center, Higashiosaka, 578-8588, Japan
| | - Yasutake Tanaka
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yoshinori Fujimura
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Hiroaki Onda
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yui Hatakeyama
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | | | | | - Wei Guo
- Dept. of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Dept. of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - J. Alan Diehl
- Dept. of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ben Z. Stanger
- Dept. of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hirofumi Tachibana
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Terence P. Gade
- Dept. of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ellen Puré
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Constantinos Koumenis
- Dept. of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Serge Y. Fuchs
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
9
|
Tian A, Meng F, Li S, Wu Y, Zhang C, Luo X. Inadequate linear catch-up growth in children born small for gestational age: Influencing factors and underlying mechanisms. Rev Endocr Metab Disord 2024; 25:805-816. [PMID: 38763958 PMCID: PMC11294269 DOI: 10.1007/s11154-024-09885-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/01/2024] [Indexed: 05/21/2024]
Abstract
A minority of children born small for gestational age (SGA) may experience catch-up growth failure and remain short in adulthood. However, the underlying causes and mechanisms of this phenomenon are not yet fully comprehended. We reviewed the present state of research concerning the growth hormone-insulin-like growth factor axis and growth plate in SGA children who fail to achieve catch-up growth. Additionally, we explored the factors influencing catch-up growth in SGA children and potential molecular mechanisms involved. Furthermore, we considered the potential benefits of supplementary nutrition, specific dietary patterns, probiotics and drug therapy in facilitating catch-up growth.
Collapse
Affiliation(s)
- Anran Tian
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fucheng Meng
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sujuan Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yichi Wu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Cai Zhang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
10
|
Tofts L, Ireland P, Tate T, Raj S, Carroll T, Munns CF, Knipe S, Langdon K, McGregor L, McKenzie F, Zankl A, Savarirayan R. Consensus Guidelines for the Use of Vosoritide in Children with Achondroplasia in Australia. CHILDREN (BASEL, SWITZERLAND) 2024; 11:789. [PMID: 39062238 PMCID: PMC11274906 DOI: 10.3390/children11070789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Achondroplasia, the most prevalent skeletal dysplasia, stems from a functional mutation in the fibroblast growth factor receptor 3 gene, leading to growth impairment. This condition presents multifaceted medical, functional and psychosocial challenges throughout childhood, adolescence and adulthood. Current management strategies aim to minimise medical complications, optimise functional capabilities and provide comprehensive supportive care. Vosoritide (trade name: VOXZOGO®, BioMarin Pharmaceuticals) is the first disease-modifying pharmaceutical treatment approved for the management of patients with achondroplasia and became available in Australia in May 2023. METHODS Standardised clinical guidelines for its optimal use are not yet widely available. To address this gap, a multidisciplinary Australian Vosoritide Working Group, comprising 12 experts with experience in achondroplasia management from across Australia, developed recommendations to guide the use of vosoritide in clinical practice. RESULTS The recommendations, which are expert opinions of the Australian Vosoritide Working Group, aim to (i) standardise the use of vosoritide across Australia, (ii) support the safe clinical rollout of vosoritide and (iii) support universal access. CONCLUSIONS These recommendations have been developed for healthcare professionals and institutions that are engaged in using vosoritide in the management of achondroplasia and will be revised using a formal framework for clinical guideline development once more evidence is available.
Collapse
Affiliation(s)
- Louise Tofts
- Health and Human Sciences, Macquarie University, Macquarie Park, Sydney, NSW 2109, Australia
| | - Penny Ireland
- Queensland Children’s Hospital, South Brisbane, QLD 4101, Australia
- School of Health and Rehabilitation Sciences, University of Queensland, St Lucia, QLD 4072, Australia
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Tracy Tate
- Kids Rehab, The Children’s Hospital at Westmead, Sydney, NSW 2145, Australia
| | - Supriya Raj
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Theresa Carroll
- Queensland Children’s Hospital, South Brisbane, QLD 4101, Australia
| | - Craig F. Munns
- Queensland Children’s Hospital, South Brisbane, QLD 4101, Australia
- Child Health Research Centre, University of Queensland, South Brisbane, QLD 4101, Australia
| | - Stephen Knipe
- The Newcastle Paediatric Clinic, New Lambton Heights, NSW 2305, Australia;
| | - Katherine Langdon
- Perth Children’s Hospital, Perth, WA 6009, Australia
- Telethon Kid’s Institute, Perth, WA 6009, Australia
| | - Lesley McGregor
- Women’s and Children’s Hospital, Adelaide, SA 5006, Australia
| | - Fiona McKenzie
- Genetic Health WA, King Edward Memorial Hospital, Perth, WA 6008, Australia
- School of Paediatrics and Child Health, University of Western Australia, Perth, WA 6009, Australia
| | - Andreas Zankl
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Department of Clinical Genetics, The Children’s Hospital at Westmead, Sydney, NSW 2145, Australia
| | - Ravi Savarirayan
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
- Murdoch Children’s Research Institute, Royal Children’s Hospital, University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
11
|
Nishioka A, Adachi N, Tanaka H, Oda Y. Two Cases of Cardiovascular Adverse Events Following Subcutaneous Vosoritide Injection in Early Infancy. Cureus 2024; 16:e59695. [PMID: 38841012 PMCID: PMC11150171 DOI: 10.7759/cureus.59695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2024] [Indexed: 06/07/2024] Open
Abstract
Achondroplasia, characterized by short stature and skeletal abnormalities, is caused by a gain-of-function variant in the fibroblast growth factor receptor 3 gene. Vosoritide, a C-type natriuretic peptide analog, is an emerging treatment for achondroplasia that functions by promoting endochondral ossification. Vosoritide was approved for the treatment of achondroplasia in Europe and the United States in 2021, and in Japan, the following year. However, vosoritide is associated with a risk of hypotension and vomiting after subcutaneous injection due to its vasodilating effect. Herein, we present two cases of cardiovascular adverse events in infants following vosoritide injection. Case 1 involved a one-month-old female infant with achondroplasia who received the first subcutaneous injection of vosoritide 30 minutes after her last formula intake. Following injection, she developed transient symptomatic hypotension accompanied by vomiting. Although established guidelines recommend that injections be administered after approximately 30 minutes (Europe/Japan) or within one hour (USA) following the last feeding, an extended interval of 1.5 to two hours was required to prevent hypotension-associated vomiting. Case 2 involved a three-month-old female infant with achondroplasia. The first subcutaneous vosoritide injection was administered four hours after the last formula intake, and she subsequently developed prolonged compensated shock with marked tachycardia requiring intervention, including repetitive bolus saline injection. These cases indicate the need to monitor patients for cardiovascular adverse events following subcutaneous injection of vosoritide in early infancy.
Collapse
Affiliation(s)
- Atsushi Nishioka
- Department of Pediatrics, Chigasaki Municipal Hospital, Chigasaki, JPN
| | - Natsuho Adachi
- Department of Pediatrics, The University of Tokyo, Tokyo, JPN
| | - Hiroyuki Tanaka
- Department of Pediatrics, The University of Tokyo, Tokyo, JPN
| | - Yoichiro Oda
- Department of Pediatrics, Chigasaki Municipal Hospital, Chigasaki, JPN
| |
Collapse
|
12
|
Jiang X, Gao L, Li Z, Shen Y, Lin ZH. Development and Challenges of Cyclic Peptides for Immunomodulation. Curr Protein Pept Sci 2024; 25:353-375. [PMID: 37990433 DOI: 10.2174/0113892037272528231030074158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 11/23/2023]
Abstract
Cyclic peptides are polypeptide chains formed by cyclic sequences of amide bonds between protein-derived or non-protein-derived amino acids. Compared to linear peptides, cyclic peptides offer several unique advantages, such as increased stability, stronger affinity, improved selectivity, and reduced toxicity. Cyclic peptide has been proved to have a promising application prospect in the medical field. In addition, this paper mainly describes that cyclic peptides play an important role in anti-cancer, anti-inflammatory, anti-virus, treatment of multiple sclerosis and membranous nephropathy through immunomodulation. In order to know more useful information about cyclic peptides in clinical research and drug application, this paper also summarizes cyclic peptides currently in the clinical trial stage and cyclic peptide drugs approved for marketing in the recent five years. Cyclic peptides have many advantages and great potential in treating various diseases, but there are still many challenges to be solved in the development process of cyclic peptides.
Collapse
Affiliation(s)
- Xianqiong Jiang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, 405400 Chongqing, China
| | - Li Gao
- School of Pharmacy and Bioengineering, Chongqing University of Technology, 405400 Chongqing, China
| | - Zhilong Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, 405400 Chongqing, China
| | - Yan Shen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, 405400 Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
- Chongqing Key Laboratory of Target Based Drug Screening and Activity Evaluation, Chongqing University of Technology, Chongqing 400054, China
| | - Zhi-Hua Lin
- School of Pharmacy and Bioengineering, Chongqing University of Technology, 405400 Chongqing, China
- Chongqing College of Traditional Chinese Medicine, 402760
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
- Chongqing Key Laboratory of Target Based Drug Screening and Activity Evaluation, Chongqing University of Technology, Chongqing 400054, China
| |
Collapse
|
13
|
Lin YW, Kao HJ, Chen WT, Kao CF, Wu JY, Chen YT, Lee YC. Cell-based screen identifies porphyrins as FGFR3 activity inhibitors with therapeutic potential for achondroplasia and cancer. JCI Insight 2023; 8:e171257. [PMID: 37824212 PMCID: PMC10721322 DOI: 10.1172/jci.insight.171257] [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: 04/06/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023] Open
Abstract
Overactive fibroblast growth factor receptor 3 (FGFR3) signaling drives pathogenesis in a variety of cancers and a spectrum of short-limbed bone dysplasias, including the most common form of human dwarfism, achondroplasia (ACH). Targeting FGFR3 activity holds great promise as a therapeutic approach for treatment of these diseases. Here, we established a receptor/adaptor translocation assay system that can specifically monitor FGFR3 activation, and we applied it to identify FGFR3 modulators from complex natural mixtures. An FGFR3-suppressing plant extract of Amaranthus viridis was identified from the screen, and 2 bioactive porphyrins, pheophorbide a (Pa) and pyropheophorbide a, were sequentially isolated from the extract and functionally characterized. Further analysis showed that Pa reduced excessive FGFR3 signaling by decreasing its half-life in FGFR3-overactivated multiple myeloma cells and chondrocytes. In an ex vivo culture system, Pa alleviated defective long bone growth in humanized ACH mice (FGFR3ACH mice). Overall, our study presents an approach to discovery and validation of plant extracts or drug candidates that target FGFR3 activation. The compounds identified by this approach may have applications as therapeutics for FGFR3-associated cancers and skeletal dysplasias.
Collapse
Affiliation(s)
- Yun-Wen Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hsiao-Jung Kao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wei-Ting Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuan-Tsong Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | - Yi-Ching Lee
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
14
|
Simran, S KDS, Dushantrao SC, Joga R, Kumar S. Vosoritide, a miracle drug, covering unmet need in achondroplasia: A regulatory update. Intractable Rare Dis Res 2023; 12:257-261. [PMID: 38024582 PMCID: PMC10680156 DOI: 10.5582/irdr.2023.01055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/02/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Dwarfism is a rare condition characterized by small stature. Achondroplasia is predominantly considered the leading cause of dwarfism. Although the condition is not life-threatening, it dramatically impacts the social life of the patient. The United States Food and Drug Administration (US FDA) first approved the drug Voxzogo (vosoritide) for achondroplasia. The drug also received approval from the European Medicines Agency (EMA) via the centralized procedure. The drug is associated with a decrease in blood pressure, a severe adverse event. However, this adverse event/risk has been overcome by benefits, i.e. fulfilling of unmet medical need. In the United States, the drug received accelerated approval as it satisfied the criteria of rare pediatric disease. This review includes a detailed orphan drug approval process with particular reference to vosoritide, which is considered a milestone for the treatment of achondroplasia.
Collapse
Affiliation(s)
- Simran
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Kirthiga Devi S S
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Sabanis Chetan Dushantrao
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Ramesh Joga
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Sandeep Kumar
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
15
|
Onesimo R, Sforza E, Bedeschi MF, Leoni C, Giorgio V, Rigante D, De Rose C, Kuczynska EM, Romeo DM, Palmacci O, Massimi L, Porro M, Gonfiantini MV, Selicorni A, Allegri A, Maghnie M, Zampino G. How pain affect real life of children and adults with achondroplasia: A systematic review. Eur J Med Genet 2023; 66:104850. [PMID: 37758167 DOI: 10.1016/j.ejmg.2023.104850] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/11/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023]
Abstract
The clinical features of achondroplasia can cause acute self-limited pain that can evolve into chronic pain. Pain causes a low quality of life, in terms of physical, emotional, social, and school functioning in both adult and children with achondroplasia. We conducted a systematic review according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) statement to describe prevalence, assessment tools, causes and management strategies of pain in this rare disease. We found that shoulder and knee pain is typically referred during infancy, while knee pain is generally referred around 5-6 years of age. The prevalence of general pain in adolescence can be as high as 90%. Chronic pain in the achondroplasia population increases with age, with up to 70% of adults reporting general pain and back pain. Recognizing the multiple determinants of acute and chronic pain in patients with achondroplasia may enable physicians to better understand and manage this burden, particularly with the advent of new drugs that may modify some of the striking features of achondroplasia.
Collapse
Affiliation(s)
- Roberta Onesimo
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy.
| | | | | | - Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy.
| | - Valentina Giorgio
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy; Università Cattolica Del Sacro Cuore, Rome, 00168, Italy.
| | - Donato Rigante
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy.
| | - Cristina De Rose
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy.
| | - Eliza Maria Kuczynska
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy.
| | - Domenico Marco Romeo
- Pediatric Neurology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168, Rome, Italy; Pediatric Neurology Unit, Università Cattolica Del Sacro Cuore, 00168, Rome, Italy.
| | - Osvaldo Palmacci
- Department of Orthopaedics and Traumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica Del Sacro Cuore, Rome, Italy.
| | - Luca Massimi
- Neurochirurgia Pediatrica, Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario A. Gemelli IRCCS, Italy; Dipartimento di Neuroscienze, Università Cattolica Del Sacro Cuore, Italy.
| | - Matteo Porro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pediatric Physical Medicine & Rehabilitation Service, Milan, Italy.
| | | | - Angelo Selicorni
- Department of Paediatrics, Presidio S. Fermo, ASST Lariana, Como, Italy.
| | - Anna Allegri
- Pediatric Endocrinology Unit, Department of Pediatrics, IRCCS IstitutoGianninaGaslini, 16147, Genoa, Italy.
| | - Mohamad Maghnie
- Pediatric Endocrinology Unit, Department of Pediatrics, IRCCS IstitutoGianninaGaslini, 16147, Genoa, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132, Genoa, Italy.
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy.
| |
Collapse
|
16
|
Rintz E, Herreño-Pachón AM, Celik B, Nidhi F, Khan S, Benincore-Flórez E, Tomatsu S. Bone Growth Induction in Mucopolysaccharidosis IVA Mouse. Int J Mol Sci 2023; 24:9890. [PMID: 37373036 PMCID: PMC10298227 DOI: 10.3390/ijms24129890] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/17/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Mucopolysaccharidosis IVA (MPS IVA; Morquio A syndrome) is caused by a deficiency of the N-acetylgalactosamine-6-sulfate-sulfatase (GALNS) enzyme, leading to the accumulation of glycosaminoglycans (GAG), keratan sulfate (KS) and chondroitin-6-sulfate (C6S), mainly in cartilage and bone. This lysosomal storage disorder (LSD) is characterized by severe systemic skeletal dysplasia. To this date, none of the treatment options for the MPS IVA patients correct bone pathology. Enzyme replacement therapy with elosulfase alpha provides a limited impact on bone growth and skeletal lesions in MPS IVA patients. To improve bone pathology, we propose a novel gene therapy with a small peptide as a growth-promoting agent for MPS IVA. A small molecule in this peptide family has been found to exert biological actions over the cardiovascular system. This work shows that an AAV vector expressing a C-type natriuretic (CNP) peptide induces bone growth in the MPS IVA mouse model. Histopathological analysis showed the induction of chondrocyte proliferation. CNP peptide also changed the pattern of GAG levels in bone and liver. These results suggest the potential for CNP peptide to be used as a treatment in MPS IVA patients.
Collapse
Affiliation(s)
- Estera Rintz
- Nemours Children’s Health, Wilmington, DE 19803, USA; (E.R.); (A.M.H.-P.); (B.C.); (F.N.); (S.K.); (E.B.-F.)
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland
| | - Angélica María Herreño-Pachón
- Nemours Children’s Health, Wilmington, DE 19803, USA; (E.R.); (A.M.H.-P.); (B.C.); (F.N.); (S.K.); (E.B.-F.)
- Faculty of Arts and Sciences, University of Delaware, Newark, DE 19716, USA
| | - Betul Celik
- Nemours Children’s Health, Wilmington, DE 19803, USA; (E.R.); (A.M.H.-P.); (B.C.); (F.N.); (S.K.); (E.B.-F.)
- Faculty of Arts and Sciences, University of Delaware, Newark, DE 19716, USA
| | - Fnu Nidhi
- Nemours Children’s Health, Wilmington, DE 19803, USA; (E.R.); (A.M.H.-P.); (B.C.); (F.N.); (S.K.); (E.B.-F.)
- Faculty of Arts and Sciences, University of Delaware, Newark, DE 19716, USA
| | - Shaukat Khan
- Nemours Children’s Health, Wilmington, DE 19803, USA; (E.R.); (A.M.H.-P.); (B.C.); (F.N.); (S.K.); (E.B.-F.)
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19144, USA
| | - Eliana Benincore-Flórez
- Nemours Children’s Health, Wilmington, DE 19803, USA; (E.R.); (A.M.H.-P.); (B.C.); (F.N.); (S.K.); (E.B.-F.)
| | - Shunji Tomatsu
- Nemours Children’s Health, Wilmington, DE 19803, USA; (E.R.); (A.M.H.-P.); (B.C.); (F.N.); (S.K.); (E.B.-F.)
- Faculty of Arts and Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19144, USA
| |
Collapse
|
17
|
Hua J, Huang J, Li G, Lin S, Cui L. Glucocorticoid induced bone disorders in children: Research progress in treatment mechanisms. Front Endocrinol (Lausanne) 2023; 14:1119427. [PMID: 37082116 PMCID: PMC10111257 DOI: 10.3389/fendo.2023.1119427] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/20/2023] [Indexed: 04/22/2023] Open
Abstract
Long-term or supra-physiological dose of glucocorticoid (GC) application in clinic can lead to impaired bone growth and osteoporosis. The side effects of GC on the skeletal system are particularly serious in growing children, potentially causing growth retardation or even osteoporotic fractures. Children's bone growth is dependent on endochondral ossification of growth plate chondrocytes, and excessive GC can hinder the development of growth plate and longitudinal bone growth. Despite the availability of drugs for treating osteoporosis, they have failed to effectively prevent or treat longitudinal bone growth and development disorders caused by GCs. As of now, there is no specific drug to mitigate these severe side effects. Traditional Chinese Medicine shows potential as an alternative to the current treatments by eliminating the side effects of GC. In summary, this article comprehensively reviews the research frontiers concerning growth and development disorders resulting from supra-physiological levels of GC and discusses the future research and treatment directions for optimizing steroid therapy. This article may also provide theoretical and experimental insight into the research and development of novel drugs to prevent GC-related side effects.
Collapse
Affiliation(s)
- Junying Hua
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Jianping Huang
- Department of Prosthodontics, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Sien Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Liao Cui
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
18
|
Gevers EF, de Winter JP. New developments and therapies in pediatric endocrinology. Eur J Pediatr 2023; 182:1439-1443. [PMID: 36567374 DOI: 10.1007/s00431-022-04772-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Evelien F Gevers
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
- Department of Paediatric Endocrinology and Diabetes, Barts Health NHS Trust - The Royal London Children's Hospital, London, United Kingdom.
| | - J Peter de Winter
- Department of Pediatrics, Spaarne Gasthuis, Haarlem/Hoofddorp, The Netherlands
- Leuven Child and Health Institute, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Cachorro E, Günscht M, Schubert M, Sadek MS, Siegert J, Dutt F, Bauermeister C, Quickert S, Berning H, Nowakowski F, Lämmle S, Firneburg R, Luo X, Künzel SR, Klapproth E, Mirtschink P, Mayr M, Dewenter M, Vettel C, Heijman J, Lorenz K, Guan K, El-Armouche A, Wagner M, Kämmerer S. CNP Promotes Antiarrhythmic Effects via Phosphodiesterase 2. Circ Res 2023; 132:400-414. [PMID: 36715019 PMCID: PMC9930893 DOI: 10.1161/circresaha.122.322031] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Ventricular arrhythmia and sudden cardiac death are the most common lethal complications after myocardial infarction. Antiarrhythmic pharmacotherapy remains a clinical challenge and novel concepts are highly desired. Here, we focus on the cardioprotective CNP (C-type natriuretic peptide) as a novel antiarrhythmic principle. We hypothesize that antiarrhythmic effects of CNP are mediated by PDE2 (phosphodiesterase 2), which has the unique property to be stimulated by cGMP to primarily hydrolyze cAMP. Thus, CNP might promote beneficial effects of PDE2-mediated negative crosstalk between cAMP and cGMP signaling pathways. METHODS To determine antiarrhythmic effects of cGMP-mediated PDE2 stimulation by CNP, we analyzed arrhythmic events and intracellular trigger mechanisms in mice in vivo, at organ level and in isolated cardiomyocytes as well as in human-induced pluripotent stem cell-derived cardiomyocytes. RESULTS In ex vivo perfused mouse hearts, CNP abrogated arrhythmia after ischemia/reperfusion injury. Upon high-dose catecholamine injections in mice, PDE2 inhibition prevented the antiarrhythmic effect of CNP. In mouse ventricular cardiomyocytes, CNP blunted the catecholamine-mediated increase in arrhythmogenic events as well as in ICaL, INaL, and Ca2+ spark frequency. Mechanistically, this was driven by reduced cellular cAMP levels and decreased phosphorylation of Ca2+ handling proteins. Key experiments were confirmed in human iPSC-derived cardiomyocytes. Accordingly, the protective CNP effects were reversed by either specific pharmacological PDE2 inhibition or cardiomyocyte-specific PDE2 deletion. CONCLUSIONS CNP shows strong PDE2-dependent antiarrhythmic effects. Consequently, the CNP-PDE2 axis represents a novel and attractive target for future antiarrhythmic strategies.
Collapse
Affiliation(s)
- Eleder Cachorro
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mario Günscht
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mario Schubert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mirna S. Sadek
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Johanna Siegert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Fabian Dutt
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Carla Bauermeister
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Susann Quickert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Henrik Berning
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Felix Nowakowski
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Simon Lämmle
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Rebecca Firneburg
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Xiaojing Luo
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Stephan R. Künzel
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Erik Klapproth
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Germany (P.M.)
| | - Manuel Mayr
- The James Black Centre, King’s College, University of London, United Kingdom (M.M.)
- Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany (M.M.)
| | - Matthias Dewenter
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Germany (M.D.)
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany (M.D., C.V.)
| | - Christiane Vettel
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany (M.D., C.V.)
- Institute of Experimental and Clinical Pharmacology and Toxicology, University Medical Center Mannheim, Germany (C.V.)
| | - Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.)
| | - Kristina Lorenz
- Institut für Pharmakologie und Toxikologie, Julius-Maximilians-Universität Würzburg, Germany (K.L.)
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany (K.L.)
| | - Kaomei Guan
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Ali El-Armouche
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Michael Wagner
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
- Bereich Rhythmologie, Klinik für Innere Medizin und Kardiologie, Herzzentrum Dresden, Dresden University of Technology, Germany (M.W.)
| | - Susanne Kämmerer
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| |
Collapse
|
20
|
Stehle D, Barresi M, Schulz J, Feil R. Heterogeneity of cGMP signalling in tumour cells and the tumour microenvironment: Challenges and chances for cancer pharmacology and therapeutics. Pharmacol Ther 2023; 242:108337. [PMID: 36623589 DOI: 10.1016/j.pharmthera.2023.108337] [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: 11/07/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/08/2023]
Abstract
The second messenger cyclic guanosine monophosphate (cGMP) is an important regulator of human (patho-)physiology and has emerged as an attractive drug target. Currently, cGMP-elevating drugs are mainly used to treat cardiovascular diseases, but there is also increasing interest in exploring their potential for cancer prevention and therapy. In this review article, we summarise recent findings in cancer-related cGMP research, with a focus on melanoma, breast cancer, colorectal cancer, prostate cancer, glioma, and ovarian cancer. These studies indicate tremendous heterogeneity of cGMP signalling in tumour tissue. It appears that different tumour and stroma cells, and perhaps different sexes, express different cGMP generators, effectors, and degraders. Therefore, the same cGMP-elevating drug can lead to different outcomes in different tumour settings, ranging from inhibition to promotion of tumourigenesis or therapy resistance. These findings, together with recent evidence that increased cGMP signalling is associated with worse prognosis in several human cancers, challenge the traditional view that cGMP elevation generally has an anti-cancer effect. As cGMP pathways appear to be more stable in the stroma than in tumour cells, we suggest that cGMP-modulating drugs should preferentially target the tumour microenvironment. Indeed, there is evidence that phosphodiesterase 5 inhibitors like sildenafil enhance anti-tumour immunity by acting on immune cells. Moreover, many in vivo results obtained with cGMP-modulating drugs could be explained by effects on the tumour vasculature rather than on the tumour cells themselves. We therefore propose a model that incorporates the NO/cGMP signalling pathway in tumour vessels as a key target for cancer therapy. Deciphering the multifaceted roles of cGMP in cancer is not only a challenge for basic research, but also provides a chance to predict potential adverse effects of cGMP-modulating drugs in cancer patients and to develop novel anti-tumour therapies by precision targeting of the relevant cells and molecular pathways.
Collapse
Affiliation(s)
- Daniel Stehle
- Interfakultäres Institut für Biochemie (IFIB), Universität Tübingen, Tübingen, Germany
| | - Mariagiovanna Barresi
- Interfakultäres Institut für Biochemie (IFIB), Universität Tübingen, Tübingen, Germany
| | - Jennifer Schulz
- Interfakultäres Institut für Biochemie (IFIB), Universität Tübingen, Tübingen, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie (IFIB), Universität Tübingen, Tübingen, Germany.
| |
Collapse
|
21
|
Nyberg M, Terzic D, Ludvigsen TP, Mark PD, Michaelsen NB, Abildstrøm SZ, Engelmann M, Richards AM, Goetze JP. Review A State of Natriuretic Peptide Deficiency. Endocr Rev 2022; 44:379-392. [PMID: 36346821 PMCID: PMC10166265 DOI: 10.1210/endrev/bnac029] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/13/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022]
Abstract
Measurement of natriuretic peptides (NPs) has proven its clinical value as biomarker, especially in the context of heart failure (HF). In contrast, a state partial NP deficiency appears integral to several conditions in which lower NP concentrations in plasma presage overt cardiometabolic disease. Here, obesity and type 2 diabetes have attracted considerable attention. Other factors - including age, sex, race, genetics, and diurnal regulation - affect the NP "armory" and may leave some individuals more prone to development of cardiovascular disease. The molecular maturation of NPs has also proven complex with highly variable O-glycosylation within the biosynthetic precursors. The relevance of this regulatory step in post-translational propeptide maturation has recently become recognized in biomarker measurement/interpretation and cardiovascular pathophysiology. An important proportion of people appear to have reduced effective net NP bioactivity in terms of receptor activation and physiological effects. The state of NP deficiency, then, both entails a potential for further biomarker development and could also offer novel pharmacological possibilities. Alleviating the state of NP deficiency before development of overt cardiometabolic disease in selected patients could be a future path for improving precision medicine.
Collapse
Affiliation(s)
| | - Dijana Terzic
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Peter D Mark
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | - A Mark Richards
- Division of Cardiology, National University Heart Centre, National University Hospital, Singapore
| | - Jens P Goetze
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
22
|
Xue C, Fu J. Progress on growth promoting therapies other than growth hormone. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:515-520. [PMID: 37202099 PMCID: PMC10264985 DOI: 10.3724/zdxbyxb-2022-0099] [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: 03/11/2022] [Accepted: 05/30/2022] [Indexed: 05/20/2023]
Abstract
Recombinant human growth hormone is a classical therapeutic drug for children with short stature. In recent years, as the mechanism of growth in children has been further explored, growth-promoting therapies other than growth hormone have made great progress. Recombinant human insulin-like growth factor (IGF)-1 is the main treatment for primary IGF-1 deficiency, and C-type natriuretic peptide (CNP) offers a therapeutic option for children with short stature due to chondrodysplasia. Growth hormone-releasing peptide analogues stimulate growth hormone release and may be used for growth-promoting therapy. In addition, gonadotropin-releasing hormone analogue (GnRHa) and aromatase inhibitors may delay the bone age in children and may be beneficial in improving final height. In this article, the research progress of growth-promoting therapies other than growth hormones is reviewed to provide more options for the clinical treatment of children with short stature.
Collapse
|
23
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
24
|
Al Shaer D, Al Musaimi O, Albericio F, de la Torre BG. 2021 FDA TIDES (Peptides and Oligonucleotides) Harvest. Pharmaceuticals (Basel) 2022; 15:ph15020222. [PMID: 35215334 PMCID: PMC8876803 DOI: 10.3390/ph15020222] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/06/2022] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
From the medical, pharmaceutical, and social perspectives, 2021 has been a year dominated by the COVID-19 pandemic. However, despite this global health crisis, the pharmaceutical industry has continued its endeavors, and 2021 could be considered an excellent year in terms of the drugs accepted by the US Food and Drug Administration (FDA). Thus, during this year, the FDA has approved 50 novel drugs, of which 36 are new chemical entities and 14 biologics. It has also authorized 10 TIDES (8 peptides, 2 oligonucleotides), in addition to 2 antibody-drug conjugates (ADCs) whose structures contain peptides. Thus, TIDES have accounted for about 24% of the approvals in the various drug categories. Importantly, this percentage has surpassed the figure in 2020 (10%), thus reflecting the remarkable success of TIDES. In this review, the approved TIDE-based drugs are analyzed on the basis of their chemical structure, medical target, mode of action, administration route, and adverse effects.
Collapse
Affiliation(s)
- Danah Al Shaer
- KRISP, School of Laboratory of Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa; (D.A.S.); (B.G.d.l.T.)
| | - Othman Al Musaimi
- Surfaces and Particle Engineering Laboratory, Department of Chemical Engineering, Imperial College London, London SW7 2AZ, UK;
| | - Fernando Albericio
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain
- Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
- Correspondence: ; Tel.: +27-614-009-144
| | - Beatriz G. de la Torre
- KRISP, School of Laboratory of Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa; (D.A.S.); (B.G.d.l.T.)
| |
Collapse
|