1
|
Si Z, Tian L, Zhou H, Lin J, Zhou J. In Vivo Interrogation of Cell-Penetrating Peptide Function: Accumulation in Tumors and the Potential as a Specific PET Probe. Bioconjug Chem 2025. [PMID: 40202497 DOI: 10.1021/acs.bioconjchem.5c00128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
We aimed to evaluate the biodistribution and specificity of 68Ga-DOTA-TAT and RHO-TAT using MGC-803 and HT-29 tumor cells as well as tumor-xenografted nude mice and to demonstrate its application in positron emission tomography (PET) imaging. The in vitro evaluation of 68Ga-DOTA-TAT was assessed in MGC-803 and HT-29 cell lines, and the in vivo evaluation of 68Ga-DOTA-TAT was also performed in mice bearing MGC-803 or HT-29 tumors, respectively. Fluorescence microscopy was also employed to evaluate the specificity of RHO-TAT in vitro in MGC-803 and HT-29 cells as well as ex vivo in tumor slices of the corresponding tumor models. The in vivo imaging differences between 68Ga-DOTA-TAT and 18F-FDG in MGC-803 and HT-29 tumors were also studied. The biodistribution and micro-PET results demonstrated significant uptake of 68Ga-DOTA-TAT in non-FDG-avid MGC-803 tumors, whereas there was negligible uptake in FDG-avid HT-29 tumors. RHO-TAT showed superior fluorescence microscopy imaging effects in MGC-803 cells and tumor slices but not in HT-29 cells and tumor slices, which were consistent with the in vivo results. 68Ga-DOTA-TAT combined with 18F-FDG can be applied noninvasively in cancers with PET imaging for potential patient selection and stratification. We demonstrated a higher binding of 68Ga-DOTA-TAT and RHO-TAT to MGC-803 cells as well as to non-FDG-avid MGC-803 xenografted tumors and a lower binding to HT-29 cells and FDG-avid xenografted tumors. These results suggest that TAT has the potential to be a ligand for targeting certain tumors.
Collapse
Affiliation(s)
- Zhan Si
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
| | - Lulu Tian
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Hongxin Zhou
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Jiasheng Lin
- Department of Nuclear Medicine, Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200237, China
| | - Jun Zhou
- Department of Nuclear Medicine, Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200237, China
| |
Collapse
|
2
|
Li Y, Du B, Yu L, Luo H, Rong H, Gao X, Yin J. Strategies and challenges of cytosolic delivery of proteins. J Drug Target 2025:1-16. [PMID: 39862226 DOI: 10.1080/1061186x.2025.2458616] [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: 12/09/2024] [Revised: 01/11/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
The cytosolic delivery of therapeutic proteins represents a promising strategy for addressing diseases caused by protein dysfunction. Despite significant advances, efficient delivery remains challenging due to barriers such as cell membrane impermeability, endosomal sequestration and protein instability. This review summarises recent progress in protein delivery systems, including physical, chemical and biological approaches, with a particular focus on strategies that enhance endosomal escape and targeting specificity. We further discuss the clinical translatability of these approaches and propose future directions for improving delivery efficiency and safety, ultimately unlocking the therapeutic potential of intracellular proteins.
Collapse
Affiliation(s)
- Yuanyuan Li
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Baojie Du
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lichao Yu
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hong Luo
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haibo Rong
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Xiangdong Gao
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Yin
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
3
|
Liao J, Zhao L, Chen H, Zhao C, Chen S, Guo X, Wang F, Liu X, Zhang X. A Bifunctional Peptide with Penetration Ability for Treating Retinal Angiogenesis via Eye Drops. Mol Pharm 2025; 22:708-720. [PMID: 39807649 DOI: 10.1021/acs.molpharmaceut.4c00683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Numerous diseases, such as diabetic retinopathy and age-related macular degeneration, can lead to retinal neovascularization, which can seriously impair the visual function and potentially result in blindness. The presence of the blood-retina barrier makes it challenging for ocularly administered drugs to penetrate physiological barriers and reach the ocular posterior segments, including the retina and choroid. Herein, we developed an innovative bifunctional peptide, Tat-C-RP7, which exhibits excellent penetration capabilities and antiangiogenic properties aimed at treating retinal neovascularization diseases. RP7 is an NRP-1 targeting peptide that blocks vascular endothelial growth factor receptor-2 (VEGFR-2) signaling and inhibits angiogenesis, while Tat facilitates the delivery of various cargoes across biological barriers, such as the blood-retina barrier. By combining these attributes, Tat-C-RP7 is anticipated to traverse ocular barriers via ocular topical administration and exert its antiangiogenic effects in the ocular posterior segment. Experimental results demonstrated that Tat-C-RP7 significantly inhibited the proliferation and migration of rat retinal microvascular endothelial cells and effectively reduced tubule formation in vitro. Its antiangiogenic activity was confirmed in zebrafish. The outstanding penetrative capabilities of FITC-labeled Tat-C-RP7 have been validated through cell uptake assays, in vitro cell barrier models, ex-vivo ocular tissues, and in vivo studies. Besides, the half-life of Tat-C-RP7 was longer than that of RP7. In an oxygen-induced retinopathy model, Tat-C-RP7 was shown to reduce the area of angiogenesis following ocular administration. Additionally, it produced no irritating effects on the eyes of rabbits. Overall, Tat-C-RP7 demonstrates excellent ocular penetrability and antiangiogenic effects and represents a promising therapeutic option for treating retinal neovascularization diseases.
Collapse
Affiliation(s)
- Jing Liao
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 Wenhua West Road, Jinan 250012, China
| | - Lin Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 Wenhua West Road, Jinan 250012, China
- Institute of Chinese Medical Sciences, University of Macau, Xurishengyin Road, Taipa, Macau 999074, China
| | - Hongyuan Chen
- Key Laboratory of Biopharmaceuticals, Postdoctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, 1263 Shunhua Road, Jinan 250098, China
- Department of General Surgery, Shandong Provincial Hospital Affiliated Shandong First Medical University, 324 Jing Wu Road, Jinan 250021, China
| | - Chunqian Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua West Road, Jinan 250012, China
| | - Shang Chen
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua West Road, Jinan 250012, China
| | - Xiuli Guo
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 Wenhua West Road, Jinan 250012, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua West Road, Jinan 250012, China
| | - Xiaoxue Liu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University, 5 Yanerdao Road, Qingdao 266000, China
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 Wenhua West Road, Jinan 250012, China
| |
Collapse
|
4
|
Liu S, Yuan F, Dong H, Zhang J, Mao X, Liu Y, Li H. PTGES3 proteolysis using the liposomal peptide-PROTAC approach. Biol Direct 2024; 19:144. [PMID: 39726032 DOI: 10.1186/s13062-024-00580-0] [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: 08/07/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the leading cause of cancer-related deaths worldwide, and the lack of effective biomarkers for early detection leads to poor therapeutic outcomes. Prostaglandin E Synthase 3 (PTGES3) is a putative prognostic marker in many solid tumors; however, its expression and biological functions in HCC have not been determined. The proteolysis-targeting chimera (PROTAC) is an established technology for targeted protein degradation. Compared to the small-molecule PROTAC, the peptide PROTAC (p-PROTAC) utilizes peptides bound to target proteins to mediate the ubiquitination and degradation of undruggable proteins. This study aimed to use the PROTAC technology to develop a PTGES3 degrader liposome complex containing a PTGES3-binding peptide and the E3 ubiquitin ligase ligand pomalidomide for regulating cell function and provide a novel pathway for treating HCC. RESULTS In this study, we demonstrated that PTGES3 is highly expressed in HCC at the transcriptional and protein levels; furthermore, PTGES3 was identified as a novel drug target that could potentially treat HCC. Hence, we developed PTGES3-PROTACs by adjusting the ligand ratio to optimize the efficacy of degradation agents. The results revealed that PTGES3-PROTAC effectively degraded PTGES3 protein and strongly weakened the HCC malignant phenotype in vitro and in vivo. CONCLUSIONS Our findings revealed that the highly selective PTGES3 proteolysis is a potential therapeutic strategy for HCC, and PTGES3 degraders PTGES3-PROTACs can be developed as safe and effective drugs for HCC treatment.
Collapse
Affiliation(s)
- Shiwei Liu
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Fukang Yuan
- Department of General Surgery, Xuzhou Central Hospital, Xuzhou, Jiangsu, 221009, China
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Hui Dong
- Fenyang College of Shanxi Medical University, Fenyang, Shanxi, 032200, China
| | - Jiaqi Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Xinyu Mao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Yangsui Liu
- Center of Hepatobiliary Pancreatic Disease, Xuzhou Central Hospital, Xuzhou, Jiangsu, 221009, China.
- Xuzhou Central Hospital Affiliated to Medical School of Southeast University, Xuzhou, Jiangsu, 221009, China.
| | - Huansong Li
- Center of Hepatobiliary Pancreatic Disease, Xuzhou Central Hospital, Xuzhou, Jiangsu, 221009, China.
- Xuzhou Central Hospital Affiliated to Medical School of Southeast University, Xuzhou, Jiangsu, 221009, China.
| |
Collapse
|
5
|
Ma Z, Li Z, Yang T, Zhao X, Zheng C, Li Y, Li Y, Guo X, Xu L, Zheng Z, Zheng H, Xiao S. A cell-penetrating NS5B-specific nanobody inhibits bovine viral diarrhea virus replication. Microb Pathog 2024; 197:107107. [PMID: 39510363 DOI: 10.1016/j.micpath.2024.107107] [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: 09/05/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Bovine viral diarrhea virus (BVDV) causes one of the significant devastating diseases for the cattle industry worldwide. The virus can cross the placenta and result in the persistent infection of the fetus, which has hampered the efficacy and the development of vaccines. Hence, efficient antiviral strategies are urgently needed. In our previous work, a specific nanobody Nb1 against nonstructural protein 5 (NS5B) was successfully isolated, and the replication of BVDV was significantly reduced in the MDBK cell line stably expressing Nb1. Nevertheless, the Nb1 protein itself cannot enter the cells autonomously, which has severely hampered the application of the Nb1. In this work, Nb1fuses with a trans-activating transduction (TAT) peptide to form the TAT-Nb1. The TAT-Nb1 was expressed in Escherichia coli, and then purified by Ni-nitrilotriacetic acid (Ni-NTA) resin. We showed that TAT successfully delivered Nb1 into the MDBK cells, and the TAT-Nb1 efficiently inhibited the replication of BVDV in a dose-and time-dependent manner. Furthermore, the recognition site of Nb1 to NS5B was identified as NS5Baa186-487 by the yeast two-hybrid assay. In summary, this study indicates that the TAT-Nb1 can potentially to be an antiviral drug against BVDV infection, and this research may accelerate the process of Nb1 for clinical use.
Collapse
Affiliation(s)
- Zhiqian Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China
| | - Zhiwei Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China
| | - Ting Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China
| | - Xiaojing Zhao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China
| | - Congsen Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China
| | - Yongqi Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China
| | - Yang Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China
| | - Xuyang Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China
| | - Lele Xu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China
| | - Zifang Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China
| | - Shuqi Xiao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, Gansu, China.
| |
Collapse
|
6
|
Obeagu EI. Exploring the role of eosinophil cationic protein (ECP) in schizophrenia: Insights and implications. Medicine (Baltimore) 2024; 103:e38380. [PMID: 39259079 PMCID: PMC11142839 DOI: 10.1097/md.0000000000038380] [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: 12/27/2023] [Revised: 03/12/2024] [Accepted: 05/06/2024] [Indexed: 09/12/2024] Open
Abstract
Schizophrenia, a multifaceted neuropsychiatric disorder characterized by disruptions in perception, cognition, and behavior, has been associated with neuroinflammatory processes. Emerging research has increasingly recognized the potential involvement of immune-related factors in the pathogenesis of schizophrenia, prompting investigations into biomarkers associated with inflammatory cascades. Among these biomarkers, Eosinophil Cationic Protein (ECP), traditionally known for its role in eosinophil-mediated immune responses, has garnered attention for its putative association with neuroinflammation in schizophrenia. This paper critically examines the current understanding of the role of ECP in schizophrenia. ECP, a cytotoxic protein released by eosinophils, has diverse immunomodulatory effects and has been identified in altered concentrations in individuals with schizophrenia. Studies have reported elevated levels of ECP in peripheral fluids of schizophrenia patients, suggesting a possible link between ECP dysregulation and the inflammatory milieu characteristic of the disorder. Moreover, the potential implications of ECP in neuroinflammatory processes relevant to schizophrenia pathophysiology are discussed. ECP's role in modulating immune responses and its potential impact on neuronal function, synaptic plasticity, and neurotoxicity within the central nervous system (CNS) are considered, highlighting the potential contribution of ECP to the neuroinflammatory mechanisms underlying schizophrenia. In conclusion, while the precise role of ECP in schizophrenia pathogenesis warrants further elucidation, exploring its association with neuroinflammation holds promise in unraveling new biomarkers and therapeutic avenues for managing this complex psychiatric disorder.
Collapse
|
7
|
Tillmanns J, Kicuntod J, Lösing J, Marschall M. 'Getting Better'-Is It a Feasible Strategy of Broad Pan-Antiherpesviral Drug Targeting by Using the Nuclear Egress-Directed Mechanism? Int J Mol Sci 2024; 25:2823. [PMID: 38474070 DOI: 10.3390/ijms25052823] [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: 01/28/2024] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The herpesviral nuclear egress represents an essential step of viral replication efficiency in host cells, as it defines the nucleocytoplasmic release of viral capsids. Due to the size limitation of the nuclear pores, viral nuclear capsids are unable to traverse the nuclear envelope without a destabilization of this natural host-specific barrier. To this end, herpesviruses evolved the regulatory nuclear egress complex (NEC), composed of a heterodimer unit of two conserved viral NEC proteins (core NEC) and a large-size extension of this complex including various viral and cellular NEC-associated proteins (multicomponent NEC). Notably, the NEC harbors the pronounced ability to oligomerize (core NEC hexamers and lattices), to multimerize into higher-order complexes, and, ultimately, to closely interact with the migrating nuclear capsids. Moreover, most, if not all, of these NEC proteins comprise regulatory modifications by phosphorylation, so that the responsible kinases, and additional enzymatic activities, are part of the multicomponent NEC. This sophisticated basis of NEC-specific structural and functional interactions offers a variety of different modes of antiviral interference by pharmacological or nonconventional inhibitors. Since the multifaceted combination of NEC activities represents a highly conserved key regulatory stage of herpesviral replication, it may provide a unique opportunity towards a broad, pan-antiherpesviral mechanism of drug targeting. This review presents an update on chances, challenges, and current achievements in the development of NEC-directed antiherpesviral strategies.
Collapse
Affiliation(s)
- Julia Tillmanns
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Jintawee Kicuntod
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Josephine Lösing
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
8
|
Zhu L, Xu J, Gan R, Xu D, Wang J, Zhou J, Ma H. Exploring peptides from toad venom for source identification by LC-MS/MS using MRM method. J Pharm Biomed Anal 2024; 239:115901. [PMID: 38091819 DOI: 10.1016/j.jpba.2023.115901] [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: 10/11/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
Toad venom is a traditional Chinese medicine (TCM) with various sources and wide-ranging preparations. Previous quality assessment studies primarily concentrated on small molecular compounds like toad dienolactones and indole alkaloids, studies on macromolecular peptides and proteins as quality assessment standards remained at the qualitative stage, lacking the development of practical and convenient quantitative methods. In this study, to explore the peptides from toad venom as a new method for identifying and evaluating its source, a complete scan of the water extract of peptides from toad venom was conducted using HPLC-Quadrupole Time-of-Flight Mass Spectrometer (Q-TOF) 5600, leading to the identification of peptides based on mass spectrometry data. Subsequently, HPLC- Quadrupole-Linear Ion Trap Mass Spectrometer (Q-Trap) 5500 employing Multiple Reaction Monitoring (MRM) mode was utilized to quantitatively analyze peptides in various sources of toad venom, followed by Partial Least Squares Discriminant Analysis (PLS-DA) to further analyze the data and evaluate the effectiveness. This study highlights the importance of exploring macromolecular substance in natural products research and provides a foundation for further studies on toad venom.
Collapse
Affiliation(s)
- Lei Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Junde Xu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Rui Gan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Dihui Xu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiaojiao Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jing Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Hongyue Ma
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
9
|
Mafi A, Mannani R, Khalilollah S, Hedayati N, Salami R, Rezaee M, Dehmordi RM, Ghorbanhosseini SS, Alimohammadi M, Akhavan-Sigari R. The Significant Role of microRNAs in Gliomas Angiogenesis: A Particular Focus on Molecular Mechanisms and Opportunities for Clinical Application. Cell Mol Neurobiol 2023; 43:3277-3299. [PMID: 37414973 PMCID: PMC11409989 DOI: 10.1007/s10571-023-01385-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/25/2023] [Indexed: 07/08/2023]
Abstract
MicroRNAs (miRNAs) are non-coding RNAs with only 20-22 nucleic acids that inhibit gene transcription and translation by binding to mRNA. MiRNAs have a diverse set of target genes and can alter most physiological processes, including cell cycle checkpoints, cell survival, and cell death mechanisms, affecting the growth, development, and invasion of various cancers, including gliomas. So optimum management of miRNA expression is essential for preserving a normal biological environment. Due to their small size, stability, and capability of specifically targeting oncogenes, miRNAs have emerged as a promising marker and new biopharmaceutical targeted therapy for glioma patients. This review focuses on the most common miRNAs associated with gliomagenesis and development by controlling glioma-determining markers such as angiogenesis. We also summarized the recent research about miRNA effects on signaling pathways, their mechanistic role and cellular targets in the development of gliomas angiogenesis. Strategies for miRNA-based therapeutic targets, as well as limitations in clinical applications, are also discussed.
Collapse
Affiliation(s)
- Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Mannani
- Department of Surgery, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Shayan Khalilollah
- Department of Neurosurgery, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Raziyeh Salami
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Malihe Rezaee
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Rohollah Mousavi Dehmordi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyedeh Sara Ghorbanhosseini
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Alimohammadi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tübingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warsaw, Poland
| |
Collapse
|
10
|
Asrorov AM, Wang H, Zhang M, Wang Y, He Y, Sharipov M, Yili A, Huang Y. Cell penetrating peptides: Highlighting points in cancer therapy. Drug Dev Res 2023; 84:1037-1071. [PMID: 37195405 DOI: 10.1002/ddr.22076] [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: 04/05/2023] [Accepted: 04/29/2023] [Indexed: 05/18/2023]
Abstract
Cell-penetrating peptides (CPPs), first identified in HIV a few decades ago, deserved great attention in the last two decades; especially to support the penetration of anticancer drug means. In the drug delivery discipline, they have been involved in various approaches from mixing with hydrophobic drugs to the use of genetically conjugated proteins. The early classification as cationic and amphipathic CPPs has been extended to a few more classes such as hydrophobic and cyclic CPPs so far. Developing potential sequences utilized almost all methods of modern science: choosing high-efficiency peptides from natural protein sequences, sequence-based comparison, amino acid substitution, obtaining chemical and/or genetic conjugations, in silico approaches, in vitro analysis, animal experiments, etc. The bottleneck effect in this discipline reveals the complications that modern science faces in drug delivery research. Most CPP-based drug delivery systems (DDSs) efficiently inhibited tumor volume and weight in mice, but only in rare cases reduced their levels and continued further processes. The integration of chemical synthesis into the development of CPPs made a significant contribution and even reached the clinical stage as a diagnostic tool. But constrained efforts still face serious problems in overcoming biobarriers to reach further achievements. In this work, we reviewed the roles of CPPs in anticancer drug delivery, focusing on their amino acid composition and sequences. As the most suitable point, we relied on significant changes in tumor volume in mice resulting from CPPs. We provide a review of individual CPPs and/or their derivatives in a separate subsection.
Collapse
Affiliation(s)
- Akmal M Asrorov
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Institute of Bioorganic Chemistry, AS of Uzbekistan, Tashkent, Uzbekistan
- Department of Natural Substances Chemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Huiyuan Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Meng Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yonghui Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yang He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Mirkomil Sharipov
- Institute of Bioorganic Chemistry, AS of Uzbekistan, Tashkent, Uzbekistan
| | - Abulimiti Yili
- The Key Laboratory of Plant Resources and Chemistry of Arid Zone, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, Xinjiang, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Zhongshan Institute for Drug Discovery, Institutes of Drug Discovery and Development, Chinese Academy of Sciences, Zhongshan, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai, China
| |
Collapse
|
11
|
Zeng Y, Shen M, Singhal A, Sevink GJA, Crone N, Boyle AL, Kros A. Enhanced Liposomal Drug Delivery Via Membrane Fusion Triggered by Dimeric Coiled-Coil Peptides. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301133. [PMID: 37199140 DOI: 10.1002/smll.202301133] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/06/2023] [Indexed: 05/19/2023]
Abstract
An ideal nanomedicine system improves the therapeutic efficacy of drugs. However, most nanomedicines enter cells via endosomal/lysosomal pathways and only a small fraction of the cargo enters the cytosol inducing therapeutic effects. To circumvent this inefficiency, alternative approaches are desired. Inspired by fusion machinery found in nature, synthetic lipidated peptide pair E4/K4 is used to induce membrane fusion previously. Peptide K4 interacts specifically with E4, and it has a lipid membrane affinity and resulting in membrane remodeling. To design efficient fusogens with multiple interactions, dimeric K4 variants are synthesized to improve fusion with E4-modified liposomes and cells. The secondary structure and self-assembly of dimers are studied; the parallel PK4 dimer forms temperature-dependent higher-order assemblies, while linear K4 dimers form tetramer-like homodimers. The structures and membrane interactions of PK4 are supported by molecular dynamics simulations. Upon addition of E4, PK4 induced the strongest coiled-coil interaction resulting in a higher liposomal delivery compared to linear dimers and monomer. Using a wide spectrum of endocytosis inhibitors, membrane fusion is found to be the main cellular uptake pathway. Doxorubicin delivery results in efficient cellular uptake and concomitant antitumor efficacy. These findings aid the development of efficient delivery systems of drugs into cells using liposome-cell fusion strategies.
Collapse
Affiliation(s)
- Ye Zeng
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Mengjie Shen
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Ankush Singhal
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Geert Jan Agur Sevink
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Niek Crone
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Aimee L Boyle
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Alexander Kros
- Dept. Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| |
Collapse
|
12
|
Gori A, Lodigiani G, Colombarolli SG, Bergamaschi G, Vitali A. Cell Penetrating Peptides: Classification, Mechanisms, Methods of Study, and Applications. ChemMedChem 2023; 18:e202300236. [PMID: 37389978 DOI: 10.1002/cmdc.202300236] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/02/2023]
Abstract
Cell-penetrating peptides (CPPs) encompass a class of peptides that possess the remarkable ability to cross cell membranes and deliver various types of cargoes, including drugs, nucleic acids, and proteins, into cells. For this reason, CPPs are largely investigated in drug delivery applications in the context of many diseases, such as cancer, diabetes, and genetic disorders. While sharing this functionality and some common structural features, such as a high content of positively charged amino acids, CPPs represent an extremely diverse group of elements, which can differentiate under many aspects. In this review, we summarize the most common characteristics of CPPs, introduce their main distinctive features, mechanistic aspects that drive their function, and outline the most widely used techniques for their structural and functional studies. We highlight current gaps and future perspectives in this field, which have the potential to significantly impact the future field of drug delivery and therapeutics.
Collapse
Affiliation(s)
- Alessandro Gori
- SCITEC - Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, Via Mario Bianco 9, 20131, Milano, Italy
| | - Giulia Lodigiani
- SCITEC - Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, Via Mario Bianco 9, 20131, Milano, Italy
| | - Stella G Colombarolli
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, L.go F. Vito 1, 00168, Roma, Italy
| | - Greta Bergamaschi
- SCITEC - Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, Via Mario Bianco 9, 20131, Milano, Italy
| | - Alberto Vitali
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, L.go F. Vito 1, 00168, Roma, Italy
| |
Collapse
|
13
|
Rowland L, Marjault HB, Karmi O, Grant D, Webb LJ, Friedler A, Nechushtai R, Elber R, Mittler R. A combination of a cell penetrating peptide and a protein translation inhibitor kills metastatic breast cancer cells. Cell Death Discov 2023; 9:325. [PMID: 37652915 PMCID: PMC10471752 DOI: 10.1038/s41420-023-01627-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023] Open
Abstract
Cell Penetrating Peptides (CPPs) are promising anticancer and antimicrobial drugs. We recently reported that a peptide derived from the human mitochondrial/ER membrane-anchored NEET protein, Nutrient Autophagy Factor 1 (NAF-1; NAF-144-67), selectively permeates and kills human metastatic epithelial breast cancer cells (MDA-MB-231), but not control epithelial cells. As cancer cells alter their phenotype during growth and metastasis, we tested whether NAF-144-67 would also be efficient in killing other human epithelial breast cancer cells that may have a different phenotype. Here we report that NAF-144-67 is efficient in killing BT-549, Hs 578T, MDA-MB-436, and MDA-MB-453 breast cancer cells, but that MDA-MB-157 cells are resistant to it. Upon closer examination, we found that MDA-MB-157 cells display a high content of intracellular vesicles and cellular protrusions, compared to MDA-MB-231 cells, that could protect them from NAF-144-67. Inhibiting the formation of intracellular vesicles and dynamics of cellular protrusions of MDA-MB-157 cells, using a protein translation inhibitor (the antibiotic Cycloheximide), rendered these cells highly susceptible to NAF-144-67, suggesting that under certain conditions, the killing effect of CPPs could be augmented when they are applied in combination with an antibiotic or chemotherapy agent. These findings could prove important for the treatment of metastatic cancers with CPPs and/or treatment combinations that include CPPs.
Collapse
Affiliation(s)
- Linda Rowland
- Department of Surgery, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center University of Missouri, 1201 Rollins Street, Columbia, MO, 65201, USA
| | - Henri-Baptiste Marjault
- Department of Surgery, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center University of Missouri, 1201 Rollins Street, Columbia, MO, 65201, USA
| | - Ola Karmi
- The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, Jerusalem, 9190401, Israel
| | - DeAna Grant
- Electron Microscopy Core Facility, University of Missouri, 0011 NextGen Precision Health Institute, 1030 Hitt Street, Columbia, MO, 65211, USA
| | - Lauren J Webb
- Department of Chemistry, The University of Texas at Austin, 2506 Speedway STOP A5300, Austin, TX, 78712, USA
| | - Assaf Friedler
- Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, Jerusalem, 9190401, Israel
| | - Rachel Nechushtai
- The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, Jerusalem, 9190401, Israel
| | - Ron Elber
- Institute for Computational Engineering and Science and Department of Chemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ron Mittler
- Department of Surgery, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center University of Missouri, 1201 Rollins Street, Columbia, MO, 65201, USA.
| |
Collapse
|
14
|
Dowdy SF, Gallagher CJ, Vitarella D, Brown J. A technology evaluation of the atypical use of a CPP-containing peptide in the formulation and performance of a clinical botulinum toxin product. Expert Opin Drug Deliv 2023; 20:1157-1166. [PMID: 37847051 DOI: 10.1080/17425247.2023.2251399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/21/2023] [Indexed: 10/18/2023]
Abstract
INTRODUCTION Cell-penetrating peptides (CPPs), are small peptides that facilitate cytosolic access and, thus, transport of therapeutic macromolecules to intracellular sites when conjugated to cargo proteins. As with all new delivery platforms, clinical development of CPP-containing therapeutics has faced considerable challenges. AREAS COVERED RTP004 is a novel, 35-amino acid, bi-CPP-containing excipient that binds noncovalently with its cargo (botulinum toxin type A) rather than conjugated as a fusion protein. An RTP004-containing neurotoxin formulation, daxibotulinumtoxinA-lanm for injection (DAXI), has recently been approved by the FDA. The formulation and pharmacological characteristics of RTP004 and the efficacy and safety of the RTP004-neurotoxin formulation are discussed. EXPERT OPINION RTP004 is a highly positively charged lysine- and arginine-rich structure that provides formulation stability, preventing self-aggregation of the cargo protein and adsorption to container surfaces. The presence of RTP004 in the formulation also appears to increase presynaptic binding of the neurotoxin, reduces post-injection diffusion, and thus facilitates an increase in the cleavage of the intracellular substrate for the botulinum toxin, likely through enhanced cellular uptake. The RTP004-neurotoxin formulation is the first CPP-containing product approved for clinical use. The potential for RTP004 to facilitate other therapeutic cargo molecules requires further research.
Collapse
Affiliation(s)
- Steven F Dowdy
- Department of Cellular and Molecular Medicine, University of California, La Jolla, CA, USA
| | | | | | - Jessica Brown
- Medical Affairs, Revance Therapeutics, Inc, Newark, CA, USA
| |
Collapse
|
15
|
Guo Y, Tang Y, Lu G, Gu J. p53 at the Crossroads between Doxorubicin-Induced Cardiotoxicity and Resistance: A Nutritional Balancing Act. Nutrients 2023; 15:nu15102259. [PMID: 37242146 DOI: 10.3390/nu15102259] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/19/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Doxorubicin (DOX) is a highly effective chemotherapeutic drug, but its long-term use can cause cardiotoxicity and drug resistance. Accumulating evidence demonstrates that p53 is directly involved in DOX toxicity and resistance. One of the primary causes for DOX resistance is the mutation or inactivation of p53. Moreover, because the non-specific activation of p53 caused by DOX can kill non-cancerous cells, p53 is a popular target for reducing toxicity. However, the reduction in DOX-induced cardiotoxicity (DIC) via p53 suppression is often at odds with the antitumor advantages of p53 reactivation. Therefore, in order to increase the effectiveness of DOX, there is an urgent need to explore p53-targeted anticancer strategies owing to the complex regulatory network and polymorphisms of the p53 gene. In this review, we summarize the role and potential mechanisms of p53 in DIC and resistance. Furthermore, we focus on the advances and challenges in applying dietary nutrients, natural products, and other pharmacological strategies to overcome DOX-induced chemoresistance and cardiotoxicity. Lastly, we present potential therapeutic strategies to address key issues in order to provide new ideas for increasing the clinical use of DOX and improving its anticancer benefits.
Collapse
Affiliation(s)
- Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| | - Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
16
|
Anselmo S, Sancataldo G, Baiamonte C, Pizzolanti G, Vetri V. Transportan 10 Induces Perturbation and Pores Formation in Giant Plasma Membrane Vesicles Derived from Cancer Liver Cells. Biomolecules 2023; 13:biom13030492. [PMID: 36979427 PMCID: PMC10046094 DOI: 10.3390/biom13030492] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Continuous progress has been made in the development of new molecules for therapeutic purposes. This is driven by the need to address several challenges such as molecular instability and biocompatibility, difficulties in crossing the plasma membrane, and the development of host resistance. In this context, cell-penetrating peptides (CPPs) constitute a promising tool for the development of new therapies due to their intrinsic ability to deliver therapeutic molecules to cells and tissues. These short peptides have gained increasing attention for applications in drug delivery as well as for their antimicrobial and anticancer activity but the general rules regulating the events involved in cellular uptake and in the following processes are still unclear. Here, we use fluorescence microscopy methods to analyze the interactions between the multifunctional peptide Transportan 10 (TP10) and the giant plasma membrane vesicles (GPMVs) derived from cancer cells. This aims to highlight the molecular mechanisms underlying functional interactions which bring its translocation across the membrane or cytotoxic mechanisms leading to membrane collapse and disruption. The Fluorescence Lifetime Imaging Microscopy (FLIM) method coupled with the phasor approach analysis proved to be the winning choice for following highly dynamic spatially heterogeneous events in real-time and highlighting aspects of such complex phenomena. Thanks to the presented approach, we were able to identify and monitor TP10 translocation into the lumen, internalization, and membrane-induced modifications depending on the peptide concentration regime.
Collapse
Affiliation(s)
- Sara Anselmo
- Dipartimento di Fisica e Chimica-Emilio Segré, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Giuseppe Sancataldo
- Dipartimento di Fisica e Chimica-Emilio Segré, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Concetta Baiamonte
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
- AteN Center-Advanced Technologies Network Center, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Giuseppe Pizzolanti
- AteN Center-Advanced Technologies Network Center, Università degli Studi di Palermo, 90128 Palermo, Italy
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", Università degli Studi di Palermo, 90127 Palermo, Italy
| | - Valeria Vetri
- Dipartimento di Fisica e Chimica-Emilio Segré, Università degli Studi di Palermo, 90128 Palermo, Italy
- AteN Center-Advanced Technologies Network Center, Università degli Studi di Palermo, 90128 Palermo, Italy
| |
Collapse
|
17
|
Targeting Transcription Factors ATF5, CEBPB and CEBPD with Cell-Penetrating Peptides to Treat Brain and Other Cancers. Cells 2023; 12:cells12040581. [PMID: 36831248 PMCID: PMC9954556 DOI: 10.3390/cells12040581] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Developing novel therapeutics often follows three steps: target identification, design of strategies to suppress target activity and drug development to implement the strategies. In this review, we recount the evidence identifying the basic leucine zipper transcription factors ATF5, CEBPB, and CEBPD as targets for brain and other malignancies. We describe strategies that exploit the structures of the three factors to create inhibitory dominant-negative (DN) mutant forms that selectively suppress growth and survival of cancer cells. We then discuss and compare four peptides (CP-DN-ATF5, Dpep, Bpep and ST101) in which DN sequences are joined with cell-penetrating domains to create drugs that pass through tissue barriers and into cells. The peptide drugs show both efficacy and safety in suppressing growth and in the survival of brain and other cancers in vivo, and ST101 is currently in clinical trials for solid tumors, including GBM. We further consider known mechanisms by which the peptides act and how these have been exploited in rationally designed combination therapies. We additionally discuss lacunae in our knowledge about the peptides that merit further research. Finally, we suggest both short- and long-term directions for creating new generations of drugs targeting ATF5, CEBPB, CEBPD, and other transcription factors for treating brain and other malignancies.
Collapse
|
18
|
TAT Nanobody Exerts Antiviral Effect against PRRSV In Vitro by Targeting Viral Nucleocapsid Protein. Int J Mol Sci 2023; 24:ijms24031905. [PMID: 36768238 PMCID: PMC9915258 DOI: 10.3390/ijms24031905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/08/2023] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is caused by the PRRS virus (PRRSV), which has brought huge economic losses to the pork industry worldwide since its first discovery in the late 1980s in North America. To date, there are no effective commercial vaccines or therapeutic drugs available for controlling the spread of PRRSV. Due to their unique advantages of high affinity and high specificity, nanobodies (Nbs) have received increasing attention in the process of disease diagnosis and treatment. Trans-activator transcription (TAT) can serve as a vector to carry specific proteins into cells by passing through cell membranes. In our previous study, a specific Nb against the PRRSV nucleocapsid (N) protein was screened using phage display technology. For this study, we developed a novel recombinant protein constituting a TAT-conjugated Nb, which we call TAT-Nb1. The target cell entry efficiency of TAT-Nb1 and its effect on PRRSV infection and replication were then investigated. Our results indicate that TAT delivered Nb1 into Marc-145 cells and porcine alveolar macrophages (PAMs) in a dose- and time-dependent manner. Furthermore, TAT-Nb1 dose-dependently suppressed PRRSV infection and replication, where this antiviral effect was independent of PRRSV strain. Co-immunoprecipitation results revealed that Nb1 efficiently interacted with the N protein of PRRSV. Taken together, the presented results suggest that TAT-Nb1 can effectively suppress PRRSV replication, and it may be considered as a new anti-PRRSV candidate drug.
Collapse
|
19
|
Zhou M, Zheng M, Zhou X, Tian S, Yang X, Ning Y, Li Y, Zhang S. The roles of connexins and gap junctions in the progression of cancer. Cell Commun Signal 2023; 21:8. [PMID: 36639804 PMCID: PMC9837928 DOI: 10.1186/s12964-022-01009-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/03/2022] [Indexed: 01/15/2023] Open
Abstract
Gap junctions (GJs), which are composed of connexins (Cxs), provide channels for direct information exchange between cells. Cx expression has a strong spatial specificity; however, its influence on cell behavior and information exchange between cells cannot be ignored. A variety of factors in organisms can modulate Cxs and subsequently trigger a series of responses that have important effects on cellular behavior. The expression and function of Cxs and the number and function of GJs are in dynamic change. Cxs have been characterized as tumor suppressors in the past, but recent studies have highlighted the critical roles of Cxs and GJs in cancer pathogenesis. The complex mechanism underlying Cx and GJ involvement in cancer development is a major obstacle to the evolution of therapy targeting Cxs. In this paper, we review the post-translational modifications of Cxs, the interactions of Cxs with several chaperone proteins, and the effects of Cxs and GJs on cancer. Video Abstract.
Collapse
Affiliation(s)
- Mingming Zhou
- Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, 300121 People’s Republic of China
| | - Xinyue Zhou
- Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Shifeng Tian
- Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, 300121 People’s Republic of China
| |
Collapse
|
20
|
Ying N, Lin X, Xie M, Zeng D. Effect of surface ligand modification on the properties of anti-tumor nanocarrier. Colloids Surf B Biointerfaces 2022; 220:112944. [DOI: 10.1016/j.colsurfb.2022.112944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/31/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022]
|
21
|
Juretić D. Designed Multifunctional Peptides for Intracellular Targets. Antibiotics (Basel) 2022; 11:antibiotics11091196. [PMID: 36139975 PMCID: PMC9495127 DOI: 10.3390/antibiotics11091196] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/25/2022] Open
Abstract
Nature’s way for bioactive peptides is to provide them with several related functions and the ability to cooperate in performing their job. Natural cell-penetrating peptides (CPP), such as penetratins, inspired the design of multifunctional constructs with CPP ability. This review focuses on known and novel peptides that can easily reach intracellular targets with little or no toxicity to mammalian cells. All peptide candidates were evaluated and ranked according to the predictions of low toxicity to mammalian cells and broad-spectrum activity. The final set of the 20 best peptide candidates contains the peptides optimized for cell-penetrating, antimicrobial, anticancer, antiviral, antifungal, and anti-inflammatory activity. Their predicted features are intrinsic disorder and the ability to acquire an amphipathic structure upon contact with membranes or nucleic acids. In conclusion, the review argues for exploring wide-spectrum multifunctionality for novel nontoxic hybrids with cell-penetrating peptides.
Collapse
Affiliation(s)
- Davor Juretić
- Mediterranean Institute for Life Sciences, 21000 Split, Croatia;
- Faculty of Science, University of Split, 21000 Split, Croatia;
| |
Collapse
|
22
|
A Peptide Inhibitor of the Human Cytomegalovirus Core Nuclear Egress Complex. Pharmaceuticals (Basel) 2022; 15:ph15091040. [PMID: 36145260 PMCID: PMC9505826 DOI: 10.3390/ph15091040] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/25/2022] Open
Abstract
The replication of human cytomegalovirus (HCMV) involves a process termed nuclear egress, which enables translocation of newly formed viral capsids from the nucleus into the cytoplasm. The HCMV core nuclear egress complex (core NEC), a heterodimer of viral proteins pUL50 and pUL53, is therefore considered a promising target for new antiviral drugs. We have recently shown that a 29-mer peptide presenting an N-terminal alpha-helical hook-like segment of pUL53, through which pUL53 interacts with pUL50, binds to pUL50 with high affinity, and inhibits the pUL50–pUL53 interaction in vitro. Here, we show that this peptide is also able to interfere with HCMV infection of cells, as well as with core NEC formation in HCMV-infected cells. As the target of the peptide, i.e., the pUL50–pUL53 interaction, is localized at the inner nuclear membrane of the cell, the peptide had to be equipped with translocation moieties that facilitate peptide uptake into the cell and the nucleus, respectively. For the resulting fusion peptide (NLS-CPP-Hook), specific cellular and nuclear uptake into HFF cells, as well as inhibition of infection with HCMV, could be demonstrated, further substantiating the HCMV core NEC as a potential antiviral target.
Collapse
|
23
|
Li W, Mu L, Zou Y, Wang W, Zhao H, Wu X, Liao S. Effect of Silkworm Pupa Protein Hydrolysates on Proliferation of Gastric Cancer Cells In Vitro. Foods 2022; 11:foods11152367. [PMID: 35954133 PMCID: PMC9368083 DOI: 10.3390/foods11152367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/25/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
The proliferation inhibition effects of the hydrolysates from silkworm pupa proteins on MGC-803 gastric cancer cells were investigated in this study. The specific morphological changes (cell membrane, cell nucleus and cytoskeleton) of cells were measured. In vitro, the proliferation of MGC-803 cells was inhibited by silkworm pupa protein hydrolysates (SPPHs) in a dose-dependent manner. The flow cytometry analysis showed that the blocking effect of SPPHs on the MGC-803 cells was mainly in the G0/G1-phase. The morphological changes, disintegration of the cytoskeleton and retardant cell cycles were probably related to the activation of apoptosis. Thus, SPPHs could be promising as a chemopreventive agent due to their ability to promote apoptosis of tumor cells.
Collapse
Affiliation(s)
- Weixin Li
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China
| | - Lixia Mu
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China
- Correspondence: ; Tel./Fax: +86-20-8723-6897
| | - Yuxiao Zou
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China
| | - Weifei Wang
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China
| | - Haifeng Zhao
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Xuli Wu
- School of Public Health, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Sentai Liao
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China
| |
Collapse
|
24
|
Relitti N, Saraswati AP, Carullo G, Papa A, Monti A, Benedetti R, Passaro E, Brogi S, Calderone V, Butini S, Gemma S, Altucci L, Campiani G, Doti N. Design and Synthesis of New Oligopeptidic Parvulin Inhibitors. ChemMedChem 2022; 17:e202200050. [PMID: 35357776 PMCID: PMC9321596 DOI: 10.1002/cmdc.202200050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/28/2022] [Indexed: 11/12/2022]
Abstract
Pin1 catalyzes the cis-trans isomerization of pThr-Pro or pSer-Pro amide bonds of different proteins involved in several physio/pathological processes. In this framework, recent research activity is directed towards the identification of new selective Pin1 inhibitors. Here, we developed a set ( 5a - p ) of peptide-based Pin1 inhibitors. Direct-binding experiments allowed the identification of the peptide-based inhibitor 5k as a potent ligand of Pin1. Notably, 5k binds Pin1 with a higher affinity compared to Pin4. The comparative analysis of molecular models of Pin1 and Pin4 with the selected compound, gave a rational explanation of the biochemical activity, and pinpointed the chemical elements that, if opportunely modified, may further improve inhibitory potency, pharmacological properties and selectivity of future peptide-based Parvulin inhibitors. Since 5k showed a limited cell penetration and no antiproliferative activity, it was conjugated to a polyarginine stretch, known to promote cell penetration of peptides, to obtain R8-5k derivative, which displayed an anti-proliferative effect on cancer cell lines compared to non-tumor cells. The effect of R8 on cell proliferation was also investigated. This work doubts the application of the R8 strategy for the development of cell penetrating antiproliferative peptides since it is not inert.
Collapse
Affiliation(s)
- Nicola Relitti
- University of Siena: Universita degli Studi di Siena, DBCF, ITALY
| | | | - Gabriele Carullo
- University of Siena: Universita degli Studi di Siena, DBCF, 2, Aldo Moro, 53100 Siena Italy, 53100, Siena, ITALY
| | - Alessandro Papa
- University of Siena: Universita degli Studi di Siena, DBCF, ITALY
| | | | - Rosaria Benedetti
- University of Campania Luigi Vanvitelli: Universita degli Studi della Campania Luigi Vanvitelli, Medicine, ITALY
| | - Eugenia Passaro
- University of Pisa Department of Pharmaceutical Sciences: Universita degli Studi di Pisa Dipartimento di Farmacia, Pharmacy, ITALY
| | - Simone Brogi
- University of Pisa Department of Pharmaceutical Sciences: Universita degli Studi di Pisa Dipartimento di Farmacia, Pharmacy, ITALY
| | - Vincenzo Calderone
- University of Pisa Department of Pharmaceutical Sciences: Universita degli Studi di Pisa Dipartimento di Farmacia, Pharmacy, ITALY
| | - Stefania Butini
- University of Siena: Universita degli Studi di Siena, DBCF, ITALY
| | - Sandra Gemma
- University of Siena: Universita degli Studi di Siena, DBCF, ITALY
| | - Lucia Altucci
- University of Campania Luigi Vanvitelli: Universita degli Studi della Campania Luigi Vanvitelli, Medicine, ITALY
| | - Giuseppe Campiani
- Universita degli Studi di Siena, Dipartimento di Biotecnologie, Via Aldo Moro 2, 53100, Siena, ITALY
| | - Nunzianna Doti
- CNR: Consiglio Nazionale delle Ricerche, Bioimaging, ITALY
| |
Collapse
|
25
|
Goto M, Ryoo I, Naffouje S, Mander S, Christov K, Wang J, Green A, Shilkaitis A, Das Gupta TK, Yamada T. Image-guided surgery with a new tumour-targeting probe improves the identification of positive margins. EBioMedicine 2022; 76:103850. [PMID: 35108666 PMCID: PMC8814381 DOI: 10.1016/j.ebiom.2022.103850] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/06/2022] [Accepted: 01/14/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Given the lack of visual discrepancy between malignant and surrounding normal tissue, current breast conserving surgery (BCS) is associated with a high re-excision rate. Due to the increasing cases of BCS, a novel method of complete tumour removal at the initial surgical resection is critically needed in the operating room to help optimize the surgical procedure and to confirm tumour-free edges. METHODS We developed a unique near-infrared (NIR) fluorescence imaging probe, ICG-p28, composed of the clinically nontoxic tumour-targeting peptide p28 and the FDA-approved NIR dye indocyanine green (ICG). ICG-p28 was characterized in vitro and evaluated in multiple breast cancer animal models with appropriate control probes. Our experimental approach with multiple-validations and -blinded procedures was designed to determine whether ICG-p28 can accurately identify tumour margins in mimicked intraoperative settings. FINDINGS The in vivo kinetics were analysed to optimize settings for potential clinical use. Xenograft tumours stably expressing iRFP as a tumour marker showed significant colocalization with ICG-p28, but not ICG alone. Image-guided surgery with ICG-p28 showed an over 6.6 × 103-fold reduction in residual normalized tumour DNA at the margin site relative to control approaches (i.e., surgery with ICG or palpation/visible inspection alone), resulting in an improved tumour recurrence rate (92% specificity) in multiple breast cancer animal models independent of the receptor expression status. ICG-p28 allowed accurate identification of tumour cells in the margin to increase the complete resection rate. INTERPRETATION Our simple and cost-effective approach has translational potential and offers a new surgical procedure that enables surgeons to intraoperatively identify tumour margins in a real-time, 3D fashion and that notably improves overall outcomes by reducing re-excision rates. FUNDING This work was supported by NIH/ National Institute of Biomedical Imaging and Bioengineering, R01EB023924.
Collapse
Affiliation(s)
- Masahide Goto
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Ingeun Ryoo
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Samer Naffouje
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA; Surgical Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Sunam Mander
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Konstantin Christov
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Jing Wang
- Department of Mathematics, Statistics and Computer Science, University of Illinois College of Liberal Arts and Sciences, IL 60607, USA
| | - Albert Green
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Anne Shilkaitis
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Tapas K Das Gupta
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Tohru Yamada
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA; Department of Bioengineering, University of Illinois College of Engineering, Chicago, IL 60607, USA.
| |
Collapse
|
26
|
Solish N, Carruthers J, Kaufman J, Rubio RG, Gross TM, Gallagher CJ. Overview of DaxibotulinumtoxinA for Injection: A Novel Formulation of Botulinum Toxin Type A. Drugs 2021; 81:2091-2101. [PMID: 34787840 PMCID: PMC8648634 DOI: 10.1007/s40265-021-01631-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/29/2022]
Abstract
Botulinum toxin type A (BoNTA) products are widely used for therapeutic and aesthetic indications, but there is a need for longer-lasting treatments that maintain symptom relief between injections and reduce the frequency of re-treatment. DaxibotulinumtoxinA for Injection (DAXI) is a novel BoNTA product containing highly purified 150-kDa core neurotoxin and is the first to be formulated with a proprietary stabilizing excipient peptide (RTP004) instead of human serum albumin. The positively charged RTP004 has been shown to enhance binding of the neurotoxin to neuronal surfaces, which may enhance the likelihood of neurotoxin internalization. DAXI produces robust, extended efficacy across both aesthetic and therapeutic indications. In an extensive glabellar lines clinical program, DAXI showed a high degree of efficacy, a consistent median time to loss of none or mild glabellar line severity of 24 weeks, and median time until return to baseline of up to 28 weeks. In adults with cervical dystonia, DAXI at 125 U and 250 U significantly improved Toronto Western Spasmodic Torticollis Rating Scale (TWSTRS) total scores, with a median duration of efficacy of 24 and 20 weeks, respectively, which compares favorably with the 12–14 weeks’ duration reported for approved BoNTA products. Overall, DAXI was well tolerated, and the consistent extended duration of effect suggests that DAXI has the potential to improve the management of both aesthetic and therapeutic conditions. Botulinum toxin is used to block the nerve signals that cause muscles to contract. Products containing botulinum toxin are commonly given by injection to treat muscle spasms (such as cervical dystonia, a painful condition where the neck muscles contract involuntarily) and for cosmetic treatment of frown lines. However, the effects of the currently approved botulinum toxin products typically wear off about 3–4 months after injection and so the injections must be repeated regularly. A new product called DAXI (DaxibotulinumtoxinA for Injection) has been developed. In this product, the botulinum toxin is formulated with a unique protein (called RTP004) that has been designed to help deliver the botulinum toxin to the nerve cells. Research suggests that the RTP004 protein in DAXI adheres the botulinum toxin to the nerves close to the injection site, potentially making its effect last longer. To date, DAXI has been studied in over 3800 patients. The studies have shown that DAXI is effective for treating neck spasms (cervical dystonia) and for reducing the appearance of frown lines. Importantly, the effects of DAXI lasted up to 6 months, which is longer than seen with other botulinum toxin products. The side effects seen with DAXI are consistent in nature and frequency with those seen with other botulinum toxin products. These findings suggest that DAXI can improve both medical and cosmetic treatments due to its longer-lasting effect.
Collapse
Affiliation(s)
| | - Jean Carruthers
- University of British Columbia and Jean Carruthers Cosmetic, Vancouver, BC, Canada
| | - Joely Kaufman
- Skin Associates of South Florida, Coral Gables, FL, USA
| | - Roman G Rubio
- Revance Therapeutics, Inc., 7555 Gateway Boulevard, Newark, CA, 94560, USA
| | - Todd M Gross
- Revance Therapeutics, Inc., 7555 Gateway Boulevard, Newark, CA, 94560, USA
| | - Conor J Gallagher
- Revance Therapeutics, Inc., 7555 Gateway Boulevard, Newark, CA, 94560, USA.
| |
Collapse
|
27
|
Wester A, Björkling F, Franzyk H. Evaluation of 1 H-Triazole-1-[ N, N'-Bis( tert-butoxycarbonyl)]carboxamidine in Solution-Phase and On-Resin Guanidinylation. J Org Chem 2021; 86:14371-14380. [PMID: 34661410 DOI: 10.1021/acs.joc.1c00994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Several guanidines and guanidinylated peptides have substantial potential as therapeutics, but efficient guanidinylation reagents are vital for easy access to these compounds. Presently, pyrazole-1-carboxamidine type reagents are commonly used in the transformations of amines into corresponding guanidines. Here, we report a comparative study of the utility of 1H-triazole-1-[N,N'-bis(tert-butoxycarbonyl)]carboxamidine, which was synthesized in two steps and readily upscaled to gram amounts. It exhibited excellent performance in solution-phase reactions, rapidly converting a set of representative aliphatic primary and unhindered secondary amines as well as aniline into the corresponding bis(tert-butoxycarbonyl)-protected guanidines. To enable a direct assessment of the reactivity of guanidinylation reagents, conversions were performed in deuterated solvents (d7-DMF or d8-THF), allowing for continuous analysis of the reaction mixtures by 1H and 13C NMR. Likewise, 1H-triazole-1-[N,N'-bis(tert-butoxycarbonyl)]carboxamidine proved to be a versatile reagent in solid-phase conversions, for example, a resin-bound test peptide (KFFKFFK) was fully guanidinylated in only 2 h by using 2 equivalents of the reagent per free amino group. Also, 1H-triazole-1-[N,N'-bis(tert-butoxycarbonyl)]carboxamidine proved capable of completely guanidinylating more sterically hindered N-terminal residues (e.g., N-methyl amino acids or a peptoid) in resin-bound peptides. Its superior reactivity and stability demonstrated under heating conditions make 1H-triazole-1-[N,N'-bis(tert-butoxycarbonyl)]carboxamidine a valuable guanidinylation reagent both in solution- and solid-phase synthesis.
Collapse
Affiliation(s)
- Anita Wester
- Center for Peptide-Based Antibiotics, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen East, Denmark
| | - Fredrik Björkling
- Center for Peptide-Based Antibiotics, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen East, Denmark
| | - Henrik Franzyk
- Center for Peptide-Based Antibiotics, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen East, Denmark
| |
Collapse
|
28
|
Vogel C, Paglia EB, Moroni LS, Demiate IM, Prestes RC, Kempka AP. Swine plasma peptides obtained using pepsin: In silico and in vitro properties and biological activities. BIOCATAL BIOTRANSFOR 2021. [DOI: 10.1080/10242422.2021.1981880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Cristine Vogel
- Department of Food Engineering and Chemical Engineering, Santa Catarina State University–UDESC, Pinhalzinho, Brazil
| | - Eduarda Baggio Paglia
- Department of Food Engineering and Chemical Engineering, Santa Catarina State University–UDESC, Pinhalzinho, Brazil
| | - Liziane Schittler Moroni
- Department of Food Engineering and Chemical Engineering, Santa Catarina State University–UDESC, Pinhalzinho, Brazil
| | - Ivo Mottin Demiate
- Department of Food Engineering, Ponta Grossa State University–UEPG, Ponta Grossa, Brazil
| | - Rosa Cristina Prestes
- Department of Technology and Food Science, Federal University of Santa Maria–UFSM, Santa Maria, Brazil
| | - Aniela Pinto Kempka
- Department of Food Engineering and Chemical Engineering, Santa Catarina State University–UDESC, Pinhalzinho, Brazil
| |
Collapse
|
29
|
Shoari A, Tooyserkani R, Tahmasebi M, Löwik DWPM. Delivery of Various Cargos into Cancer Cells and Tissues via Cell-Penetrating Peptides: A Review of the Last Decade. Pharmaceutics 2021; 13:1391. [PMID: 34575464 PMCID: PMC8470549 DOI: 10.3390/pharmaceutics13091391] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 01/03/2023] Open
Abstract
Cell-penetrating peptides (CPPs), also known as protein transduction domains, are a class of diverse amino acid sequences with the ability to cross cellular membranes. CPPs can deliver several bioactive cargos, including proteins, peptides, nucleic acids and chemotherapeutics, into cells. Ever since their discovery, synthetic and natural CPPs have been utilized in therapeutics delivery, gene editing and cell imaging in fundamental research and clinical experiments. Over the years, CPPs have gained significant attention due to their low cytotoxicity and high transduction efficacy. In the last decade, multiple investigations demonstrated the potential of CPPs as carriers for the delivery of therapeutics to treat various types of cancer. Besides their remarkable efficacy owing to fast and efficient delivery, a crucial benefit of CPP-based cancer treatments is delivering anticancer agents selectively, rather than mediating toxicities toward normal tissues. To obtain a higher therapeutic index and to improve cell and tissue selectivity, CPP-cargo constructions can also be complexed with other agents such as nanocarriers and liposomes to obtain encouraging outcomes. This review summarizes various types of CPPs conjugated to anticancer cargos. Furthermore, we present a brief history of CPP utilization as delivery systems for anticancer agents in the last decade and evaluate several reports on the applications of CPPs in basic research and preclinical studies.
Collapse
Affiliation(s)
- Alireza Shoari
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-111, Iran; (A.S.); (R.T.); (M.T.)
- Bio-Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Raheleh Tooyserkani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-111, Iran; (A.S.); (R.T.); (M.T.)
- Bio-Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Mehdi Tahmasebi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-111, Iran; (A.S.); (R.T.); (M.T.)
| | - Dennis W. P. M. Löwik
- Bio-Organic Chemistry, Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| |
Collapse
|
30
|
Szabo I, Zoratti M, Biasutto L. Targeting mitochondrial ion channels for cancer therapy. Redox Biol 2021; 42:101846. [PMID: 33419703 PMCID: PMC8113036 DOI: 10.1016/j.redox.2020.101846] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Pharmacological targeting of mitochondrial ion channels is emerging as a promising approach to eliminate cancer cells; as most of these channels are differentially expressed and/or regulated in cancer cells in comparison to healthy ones, this strategy may selectively eliminate the former. Perturbation of ion fluxes across the outer and inner membranes is linked to alterations of redox state, membrane potential and bioenergetic efficiency. This leads to indirect modulation of oxidative phosphorylation, which is/may be fundamental for both cancer and cancer stem cell survival. Furthermore, given the crucial contribution of mitochondria to intrinsic apoptosis, modulation of their ion channels leading to cytochrome c release may be of great advantage in case of resistance to drugs triggering apoptotic events upstream of the mitochondrial phase. In the present review, we give an overview of the known mitochondrial ion channels and of their modulators capable of killing cancer cells. In addition, we discuss state-of-the-art strategies using mitochondriotropic drugs or peptide-based approaches allowing a more efficient and selective targeting of mitochondrial ion channel-linked events.
Collapse
Affiliation(s)
- Ildiko Szabo
- Department of Biology, University of Padova, Italy; CNR Institute of Neurosciences, Padova, Italy.
| | | | - Lucia Biasutto
- CNR Institute of Neurosciences, Padova, Italy; Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
31
|
|
32
|
Kong X, Xu J, Yang X, Zhai Y, Ji J, Zhai G. Progress in tumour-targeted drug delivery based on cell-penetrating peptides. J Drug Target 2021; 30:46-60. [PMID: 33944641 DOI: 10.1080/1061186x.2021.1920026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Since the discovery of cell-penetrating peptides (CPP) in the 1980s, they have played a unique role in various fields owing to their excellent and unique cell membrane penetration function. In particular, in the treatment of tumours, CPPS have been used to deliver several types of 'cargos' to cancer cells. To address the insufficient targeting ability, non-selectivity, and blood instability, activatable cell-penetrating peptides, which can achieve targeted drug delivery in tumour treatment, enhance curative effects, and reduce toxicity have been developed. This study reviews the application of different cell-penetrating peptides in tumour-targeted delivery, overcoming multidrug resistance, organelle targeting, tumour imaging, and diagnosis, and summarises the different mechanisms of activatable cell-penetrating peptides in detail.
Collapse
Affiliation(s)
- Xinru Kong
- Key Laboratory of Chemical Biology, Department of Pharmaceutics, School of Pharmaceutical Sciences, Ministry of Education, Shandong University, Jinan, China
| | - Jiangkang Xu
- Key Laboratory of Chemical Biology, Department of Pharmaceutics, School of Pharmaceutical Sciences, Ministry of Education, Shandong University, Jinan, China
| | - Xiaoye Yang
- Key Laboratory of Chemical Biology, Department of Pharmaceutics, School of Pharmaceutical Sciences, Ministry of Education, Shandong University, Jinan, China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
| | - Jianbo Ji
- Key Laboratory of Chemical Biology, Department of Pharmaceutics, School of Pharmaceutical Sciences, Ministry of Education, Shandong University, Jinan, China
| | - Guangxi Zhai
- Key Laboratory of Chemical Biology, Department of Pharmaceutics, School of Pharmaceutical Sciences, Ministry of Education, Shandong University, Jinan, China
| |
Collapse
|
33
|
Ayad C, Libeau P, Lacroix-Gimon C, Ladavière C, Verrier B. LipoParticles: Lipid-Coated PLA Nanoparticles Enhanced In Vitro mRNA Transfection Compared to Liposomes. Pharmaceutics 2021; 13:377. [PMID: 33809164 PMCID: PMC7999670 DOI: 10.3390/pharmaceutics13030377] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
The approval of two mRNA vaccines as urgent prophylactic treatments against Covid-19 made them a realistic alternative to conventional vaccination methods. However, naked mRNA is rapidly degraded by the body and cannot effectively penetrate cells. Vectors capable of addressing these issues while allowing endosomal escape are therefore needed. To date, the most widely used vectors for this purpose have been lipid-based vectors. Thus, we have designed an innovative vector called LipoParticles (LP) consisting of poly(lactic) acid (PLA) nanoparticles coated with a 15/85 mol/mol DSPC/DOTAP lipid membrane. An in vitro investigation was carried out to examine whether the incorporation of a solid core offered added value compared to liposomes alone. To that end, a formulation strategy that we have named particulate layer-by-layer (pLbL) was used. This method permitted the adsorption of nucleic acids on the surface of LP (mainly by means of electrostatic interactions through the addition of LAH4-L1 peptide), allowing both cellular penetration and endosomal escape. After a thorough characterization of size, size distribution, and surface charge- and a complexation assessment of each vector-their transfection capacity and cytotoxicity (on antigenic presenting cells, namely DC2.4, and epithelial HeLa cells) were compared. LP have been shown to be significantly better transfecting agents than liposomes through pLbL formulation on both HeLa and DC 2.4 cells. These data illustrate the added value of a solid particulate core inside a lipid membrane, which is expected to rigidify the final assemblies and makes them less prone to early loss of mRNA. In addition, this assembly promoted not only efficient delivery of mRNA, but also of plasmid DNA, making it a versatile nucleic acid carrier that could be used for various vaccine applications. Finally, if the addition of the LAH4-L1 peptide systematically leads to toxicity of the pLbL formulation on DC 2.4 cells, the optimization of the nucleic acid/LAH4-L1 peptide mass ratio becomes an interesting strategy-essentially reducing the peptide intake to limit its cytotoxicity while maintaining a relevant transfection efficiency.
Collapse
Affiliation(s)
- Camille Ayad
- UMR 5305: Laboratoire de Biologie Tissulaire et d’Ingénierie Thérapeutique, Institut de Biologie et Chimie des Protéines, CNRS/Université Claude Bernard Lyon 1, 7 passage du Vercors, CEDEX 07, 69367 Lyon, France; (P.L.); (C.L.-G.)
| | - Pierre Libeau
- UMR 5305: Laboratoire de Biologie Tissulaire et d’Ingénierie Thérapeutique, Institut de Biologie et Chimie des Protéines, CNRS/Université Claude Bernard Lyon 1, 7 passage du Vercors, CEDEX 07, 69367 Lyon, France; (P.L.); (C.L.-G.)
| | - Céline Lacroix-Gimon
- UMR 5305: Laboratoire de Biologie Tissulaire et d’Ingénierie Thérapeutique, Institut de Biologie et Chimie des Protéines, CNRS/Université Claude Bernard Lyon 1, 7 passage du Vercors, CEDEX 07, 69367 Lyon, France; (P.L.); (C.L.-G.)
| | - Catherine Ladavière
- UMR 5223: Ingénierie des Matériaux Polymères, CNRS/Université Claude Bernard Lyon 1, Domaine Scientifique de la Doua, Bâtiment POLYTECH, 15 bd André Latarjet, CEDEX, 69622 Villeurbanne, France
| | - Bernard Verrier
- UMR 5305: Laboratoire de Biologie Tissulaire et d’Ingénierie Thérapeutique, Institut de Biologie et Chimie des Protéines, CNRS/Université Claude Bernard Lyon 1, 7 passage du Vercors, CEDEX 07, 69367 Lyon, France; (P.L.); (C.L.-G.)
| |
Collapse
|
34
|
Wang Q, Cheng S, Qin F, Fu A, Fu C. Application progress of RVG peptides to facilitate the delivery of therapeutic agents into the central nervous system. RSC Adv 2021; 11:8505-8515. [PMID: 35423368 PMCID: PMC8695342 DOI: 10.1039/d1ra00550b] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 12/14/2022] Open
Abstract
The incidence of central nervous system (CNS) diseases is increasing with the aging population. However, it remains challenging to deliver drugs into the CNS because of the existence of a blood-brain barrier (BBB). Notably, rabies virus glycoprotein (RVG) peptides have been developed as delivery ligands for CNS diseases. So far, massive RVG peptide modified carriers have been reported, such as liposomes, micelles, polymers, exosomes, dendrimers, and proteins. Moreover, these drug delivery systems can encapsulate almost all small molecules and macromolecule drugs, including siRNA, microRNAs, DNA, proteins, and other nanoparticles, to treat various CNS diseases with efficient and safe drugs. In this review, targeted delivery systems with RVG peptide modified carriers possessing favorable biocompatibility and delivery efficiency are summarized.
Collapse
Affiliation(s)
- Qinghua Wang
- Immunology Research Center of Medical Research Institute, College of Animal Medicine, Southwest University Chongqing 402460 China
| | - Shang Cheng
- Animal Husbandry Technology, Popularization Master Station of Chongqing Chongqing 401121 China
| | - Fen Qin
- The Ninth People's Hospital of Chongqing Chongqing 400702 China
| | - Ailing Fu
- College of Pharmaceutical Science, Southwest University Chongqing 400715 China +86-23-68251225 +86-23-68251225
| | - Chen Fu
- College of Pharmaceutical Science, Southwest University Chongqing 400715 China +86-23-68251225 +86-23-68251225
| |
Collapse
|
35
|
Parrasia S, Rossa A, Varanita T, Checchetto V, De Lorenzi R, Zoratti M, Paradisi C, Ruzza P, Mattarei A, Szabò I, Biasutto L. An Angiopep2-PAPTP Construct Overcomes the Blood-Brain Barrier. New Perspectives against Brain Tumors. Pharmaceuticals (Basel) 2021; 14:ph14020129. [PMID: 33562146 PMCID: PMC7914648 DOI: 10.3390/ph14020129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/30/2021] [Accepted: 02/03/2021] [Indexed: 12/23/2022] Open
Abstract
A developing family of chemotherapeutics—derived from 5-(4-phenoxybutoxy)psoralen (PAP-1)—target mitochondrial potassium channel mtKv1.3 to selectively induce oxidative stress and death of diseased cells. The key to their effectiveness is the presence of a positively charged triphenylphosphonium group which drives their accumulation in the organelles. These compounds have proven their preclinical worth in murine models of cancers such as melanoma and pancreatic adenocarcinoma. In in vitro experiments they also efficiently killed glioblastoma cells, but in vivo they were powerless against orthotopic glioma because they were completely unable to overcome the blood-brain barrier. In an effort to improve brain delivery we have now coupled one of these promising compounds, PAPTP, to well-known cell-penetrating and brain-targeting peptides TAT48–61 and Angiopep-2. Coupling has been obtained by linking one of the phenyl groups of the triphenylphosphonium to the first amino acid of the peptide via a reversible carbamate ester bond. Both TAT48–61 and Angiopep-2 allowed the delivery of 0.3–0.4 nmoles of construct per gram of brain tissue upon intravenous (i.v.) injection of 5 µmoles/kg bw to mice. This is the first evidence of PAPTP delivery to the brain; the chemical strategy described here opens the possibility to conjugate PAPTP to small peptides in order to fine-tune tissue distribution of this interesting compound.
Collapse
Affiliation(s)
- Sofia Parrasia
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy; (S.P.); (M.Z.)
| | - Andrea Rossa
- Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35131 Padova, Italy; (A.R.); (R.D.L.); (C.P.); (P.R.)
| | - Tatiana Varanita
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy; (T.V.); (V.C.); (I.S.)
| | - Vanessa Checchetto
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy; (T.V.); (V.C.); (I.S.)
| | - Riccardo De Lorenzi
- Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35131 Padova, Italy; (A.R.); (R.D.L.); (C.P.); (P.R.)
- CNR Institute of Biomolecular Chemistry, Via F. Marzolo 1, 35131 Padova, Italy
| | - Mario Zoratti
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy; (S.P.); (M.Z.)
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy
| | - Cristina Paradisi
- Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35131 Padova, Italy; (A.R.); (R.D.L.); (C.P.); (P.R.)
| | - Paolo Ruzza
- Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35131 Padova, Italy; (A.R.); (R.D.L.); (C.P.); (P.R.)
- CNR Institute of Biomolecular Chemistry, Via F. Marzolo 1, 35131 Padova, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Ildikò Szabò
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy; (T.V.); (V.C.); (I.S.)
| | - Lucia Biasutto
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy; (S.P.); (M.Z.)
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy
- Correspondence:
| |
Collapse
|
36
|
Moody TW, Lee L, Ramos-Alvarez I, Iordanskaia T, Mantey SA, Jensen RT. Bombesin Receptor Family Activation and CNS/Neural Tumors: Review of Evidence Supporting Possible Role for Novel Targeted Therapy. Front Endocrinol (Lausanne) 2021; 12:728088. [PMID: 34539578 PMCID: PMC8441013 DOI: 10.3389/fendo.2021.728088] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) are increasingly being considered as possible therapeutic targets in cancers. Activation of GPCR on tumors can have prominent growth effects, and GPCRs are frequently over-/ectopically expressed on tumors and thus can be used for targeted therapy. CNS/neural tumors are receiving increasing attention using this approach. Gliomas are the most frequent primary malignant brain/CNS tumor with glioblastoma having a 10-year survival <1%; neuroblastomas are the most common extracranial solid tumor in children with long-term survival<40%, and medulloblastomas are less common, but one subgroup has a 5-year survival <60%. Thus, there is an increased need for more effective treatments of these tumors. The Bombesin-receptor family (BnRs) is one of the GPCRs that are most frequently over/ectopically expressed by common tumors and is receiving particular attention as a possible therapeutic target in several tumors, particularly in prostate, breast, and lung cancer. We review in this paper evidence suggesting why a similar approach in some CNS/neural tumors (gliomas, neuroblastomas, medulloblastomas) should also be considered.
Collapse
Affiliation(s)
- Terry W. Moody
- Department of Health and Human Services, National Cancer Institute, Center for Cancer Training, Office of the Director, Bethesda, MD, United States
| | - Lingaku Lee
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- Department of Gastroenterology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Irene Ramos-Alvarez
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Tatiana Iordanskaia
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Samuel A. Mantey
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Robert T. Jensen
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Robert T. Jensen,
| |
Collapse
|