1
|
Sagar B, Gupta S, Verma SK, Reddy YVM, Shukla S. Navigating cancer therapy: Harnessing the power of peptide-drug conjugates as precision delivery vehicles. Eur J Med Chem 2024; 283:117131. [PMID: 39647418 DOI: 10.1016/j.ejmech.2024.117131] [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: 01/30/2024] [Revised: 11/17/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
Cancer treatment is a formidable challenge due to the adverse effects associated with non-selective therapies like chemotherapy and radiotherapy. This review article primarily centers on the application of Peptide-Drug Conjugates (PDCs) for delivering cancer treatment. PDCs represent a promising class of precision medicines, harnessing the unique attributes of peptides in conjunction with non-peptide components. The covalent linking of peptides and drugs through specialized connectors characterizes PDCs. These constructs play a pivotal role in delivering drugs directly to tumor sites with high precision. PDCs encompass three pivotal components: a targeting ligand, a cytotoxic ligand, and a carefully chosen linker. The selection of these elements is crucial to maximize the efficiency of PDCs. PDCs offer a multitude of advantages over conventional drug molecules, including enhanced specificity, reduced off-target effects, and an improved therapeutic profile. The peptide component within PDCs can be customized to specifically adhere to disease-specific receptors or biomarkers, facilitating targeted drug delivery and accumulation in afflicted cells or tissues. This targeted approach enables the controlled release of therapeutic payloads at the localized site, resulting in heightened effectiveness and minimized systemic toxicity. Diverse linker strategies are employed to ensure the stable connection between the peptide and non-peptide components, ensuring controlled drug release at the desired location of action. The peptides utilized in these treatments encompass cell-penetrating peptides, peptides designed to target tumor cells, and those aimed at the nucleus of cancer cells. While certain clinical trials have been conducted, and some PDCs are currently in use for cancer treatment, it's essential to acknowledge that PDCs have their limitations, such as low stability in plasma, fast elimination and limited oral bioavailability. Ongoing research endeavors seek to surmount these challenges and further establish PDCs as potent agents for cancer treatment. This review sheds light on recent advancements in the design, delivery, and applications of PDCs, while also highlighting the prevailing challenges and charting a path for future research directions.
Collapse
Affiliation(s)
- Bulbul Sagar
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016, New Delhi, India
| | - Sarthak Gupta
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016, New Delhi, India
| | - Sarvesh Kumar Verma
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur, 302017, Rajasthan, India
| | | | - Shefali Shukla
- Sri Venkateswara College, University of Delhi, New Delhi, India.
| |
Collapse
|
2
|
Fu X, Li J, Chen X, Chen H, Wang Z, Qiu F, Xie D, Huang J, Yue S, Cao C, Liang Y, Lu A, Liang C. Repurposing AS1411 for constructing ANM-PROTACs. Cell Chem Biol 2024; 31:1290-1304.e7. [PMID: 38657608 DOI: 10.1016/j.chembiol.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/29/2023] [Accepted: 03/29/2024] [Indexed: 04/26/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) are heterobifunctional molecules consisting of two ligands joined by a linker, enabling them to simultaneously bind with an E3 ligase and a protein of interest (POI) and trigger proteasomal degradation of the POI. Limitations of PROTAC include lack of potent E3 ligands, poor cell selectivity, and low permeability. AS1411 is an antitumor aptamer specifically recognizing a membrane-nucleus shuttling nucleolin (NCL). Here, we repurpose AS1411 as a ligand for an E3 ligase mouse double minute 2 homolog (MDM2) via anchoring the NCL-MDM2 complex. Then, we construct an AS1411-NCL-MDM2-based PROTAC (ANM-PROTAC) by conjugating AS1411 with large-molecular-weight ligands for "undruggable" oncogenic STAT3, c-Myc, p53-R175H, and AR-V7. We show that the ANM-PROTAC efficiently penetrates tumor cells, recruits MDM2 and degrades the POIs. The ANM-PROTAC achieves tumor-selective distribution and exhibits excellent antitumor activity with no systemic toxicity. This is a PROTAC with built-in tumor-targeting and cell-penetrating capacities.
Collapse
Affiliation(s)
- Xuekun Fu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Jin Li
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xinxin Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hongzhen Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Jie Huang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Siran Yue
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Chunhao Cao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Yiying Liang
- Shenzhen LingGene Biotech Co., Ltd, Shenzhen 518055, China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510006, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China.
| |
Collapse
|
3
|
Wibel R, van Hoogevest P, Drescher S. The role of phospholipids in drug delivery formulations - Recent advances presented at the Researcher's Day 2023 Conference of the Phospholipid Research Center Heidelberg. Eur J Pharm Biopharm 2024; 197:114215. [PMID: 38350530 DOI: 10.1016/j.ejpb.2024.114215] [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: 01/02/2024] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/15/2024]
Abstract
This Focus on Meetings contribution summarizes recent advances in the research on phospholipids and their applications for drug delivery and analytical purposes that have been presented at the hybrid Researcher's Day 2023 Conference of the Phospholipid Research Center (PRC), held on July 3-5, 2023, in Bad Dürkheim, Germany. The PRC is a non-profit organization focused on expanding and sharing scientific and technological knowledge of phospholipids in pharmaceutical and other applications. This is accomplished by, e.g., funding doctoral and postdoctoral research projects. The progress made with these projects is presented at the Researcher's Day Conference every two years. Four main topics were presented and discussed in various lectures: (1) formulation of phospholipid-based nanocarriers, (2) therapeutic applications of phospholipids and phospholipid-based nanocarriers, (3) phospholipids as excipients in oral, dermal, and parenteral dosage forms, and (4) interactions of phospholipids and phospholipid-based vesicles in biological environment and their use as analytical platforms.
Collapse
Affiliation(s)
- Richard Wibel
- Phospholipid Research Center, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Peter van Hoogevest
- Phospholipid Research Center, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Simon Drescher
- Phospholipid Research Center, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany.
| |
Collapse
|
4
|
Dzhumashev D, Anton-Joseph S, Morel VJ, Timpanaro A, Bordon G, Piccand C, Aleandri S, Luciani P, Rössler J, Bernasconi M. Rapid liposomal formulation for nucleolin targeting to rhabdomyosarcoma cells. Eur J Pharm Biopharm 2024; 194:49-61. [PMID: 38029941 DOI: 10.1016/j.ejpb.2023.11.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma. More effective and less toxic therapies are urgently needed for high-risk patients. Peptide-guided targeted drug delivery can increase the therapeutic index of encapsulated drugs and improve patients' well-being. To apply this strategy to RMS, we identified the peptide F3 in a screening for peptides binding to RMS cells surface. F3 binds to nucleolin, which is present on the surface of RMS cells and is abundantly expressed at the mRNA level in RMS patients' biopsies compared to healthy tissues. We developed a rapid microfluidic formulation of F3-decorated PEGylated liposomes and remote loading of the chemotherapeutic drug vincristine. Size, surface charge, drug loading and retention of targeted and control liposomes were studied. Enhanced cellular binding and uptake were observed in three different nucleolin-positive RMS cell lines. Importantly, F3-functionalized liposomes loaded with vincristine were up to 11 times more cytotoxic than non-targeted liposomes for RMS cell lines. These results demonstrate that F3-functionalized liposomes are promising for targeted drug delivery to RMS and warrant further in vivo investigations.
Collapse
Affiliation(s)
- Dzhangar Dzhumashev
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Stenija Anton-Joseph
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Victoria J Morel
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Andrea Timpanaro
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Gregor Bordon
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Caroline Piccand
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland.
| |
Collapse
|
5
|
Kumar Sharma A, Sharma R, Dev Sarma H, Mukherjee A, Das T, Satpati D. Design, synthesis and evaluation of 177Lu-labeled inverso and retro-inverso A9 peptide variants targeting HER2-overexpression. Bioorg Chem 2023; 140:106761. [PMID: 37586132 DOI: 10.1016/j.bioorg.2023.106761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 08/18/2023]
Abstract
Several HER2-specific peptides are being continuously explored to find a candidate with suitable pharmacokinetic properties for development of effective radiopharmaceutical that can find applications for clinical screening of breast cancer patients. In the present work with an aim of preparing a radiopeptide with improved metabolic stability and in vivo pharmacokinetic performance we modified our previously reported [177Lu]DOTA-L-A9 peptide. Here we designed an 'inverso' peptide with all d-amino acids and a 'retro-inverso' peptide where sequence of d-amino acids was reversed. Higher secondary structure stabilization of retro- inverso A9 variant compared to inverso A9 peptide was evident by circular dichroism studies. The two radiopeptides [177Lu]DOTA-D-A9 and [177Lu]DOTA-rD-A9 exhibited significantly improved in vivo metabolic stability over the original l-peptide. The retro-inverso variant, [177Lu]DOTA-rD-A9 demonstrated better pharmacokinetic behavior with significantly higher tumor uptake than the inverso peptide, [177Lu]DOTA-D-A9 and the original peptide, [177Lu]DOTA-L-A9. In the present case of A9 peptide, reversal of the peptide sequence of d-amino acids boosted the uptake and retention of radioactivity in HER2-positive tumor. The present study can thus guide the design and development of newer and improved versions of peptides.
Collapse
Affiliation(s)
- Amit Kumar Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India; Homi Bhabha National Institute, Mumbai, India
| | - Rohit Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India; Homi Bhabha National Institute, Mumbai, India
| | - Haladhar Dev Sarma
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India; Homi Bhabha National Institute, Mumbai, India
| | - Tapas Das
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India; Homi Bhabha National Institute, Mumbai, India
| | - Drishty Satpati
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India; Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
6
|
Yang S, Wang M, Wang T, Sun M, Huang H, Shi X, Duan S, Wu Y, Zhu J, Liu F. Self-assembled short peptides: Recent advances and strategies for potential pharmaceutical applications. Mater Today Bio 2023; 20:100644. [PMID: 37214549 PMCID: PMC10199221 DOI: 10.1016/j.mtbio.2023.100644] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/10/2023] [Accepted: 04/23/2023] [Indexed: 05/24/2023] Open
Abstract
Self-assembled short peptides have intrigued scientists due to the convenience of synthesis, good biocompatibility, low toxicity, inherent biodegradability and fast response to change in the physiological environment. Therefore, it is necessary to present a comprehensive summary of the recent advances in the last decade regarding the construction, route of administration and application of self-assembled short peptides based on the knowledge on their unique and specific ability of self-assembly. Herein, we firstly explored the molecular mechanisms of self-assembly of short peptides, such as non-modified amino acids, as well as Fmoc-modified, N-functionalized, and C-functionalized peptides. Next, cell penetration, fusion, and peptide targeting in peptide-based drug delivery were characterized. Then, the common administration routes and the potential pharmaceutical applications (drug delivery, antibacterial activity, stabilizers, imaging agents, and applications in bioengineering) of peptide drugs were respectively summarized. Last but not least, some general conclusions and future perspectives in the relevant fields were briefly listed. Although with certain challenges, great opportunities are offered by self-assembled short peptides to the fascinating area of drug development.
Collapse
Affiliation(s)
- Shihua Yang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
- Department of Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110102, China
| | - Mingge Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Tianye Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
- Department of Anus and Intestine Surgery, The First Hospital of Dalian Medical University, Dalian, 116000, China
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hanwei Huang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
- Department of Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110102, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Shijie Duan
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
- Department of Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110102, China
| | - Ying Wu
- Department of Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110102, China
| | - Jiaming Zhu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
- Department of Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110102, China
| |
Collapse
|
7
|
Functional Peptides from One-bead One-compound High-throughput Screening Technique. Chem Res Chin Univ 2023. [DOI: 10.1007/s40242-023-2356-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
8
|
Wu M, Huang W, Yang N, Liu Y. Learn from antibody–drug conjugates: consideration in the future construction of peptide-drug conjugates for cancer therapy. Exp Hematol Oncol 2022; 11:93. [DOI: 10.1186/s40164-022-00347-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractCancer is one of the leading causes of death worldwide due to high heterogeneity. Although chemotherapy remains the mainstay of cancer therapy, non-selective toxicity and drug resistance of mono-chemotherapy incur broad criticisms. Subsequently, various combination strategies have been developed to improve clinical efficacy, also known as cocktail therapy. However, conventional “cocktail administration” is just passable, due to the potential toxicities to normal tissues and unsatisfactory synergistic effects, especially for the combined drugs with different pharmacokinetic properties. The drug conjugates through coupling the conventional chemotherapeutics to a carrier (such as antibody and peptide) provide an alternative strategy to improve therapeutic efficacy and simultaneously reduce the unspecific toxicities, by virtue of the advantages of highly specific targeting ability and potent killing effect. Although 14 antibody–drug conjugates (ADCs) have been approved worldwide and more are being investigated in clinical trials so far, several limitations have been disclosed during clinical application. Compared with ADCs, peptide-drug conjugates (PDCs) possess several advantages, including easy industrial synthesis, low cost, high tissue penetration and fast clearance. So far, only a handful of PDCs have been approved, highlighting tremendous development potential. Herein, we discuss the progress and pitfalls in the development of ADCs and underline what can learn from ADCs for the better construction of PDCs in the future.
Collapse
|
9
|
Dzhumashev D, Timpanaro A, Ali S, De Micheli AJ, Mamchaoui K, Cascone I, Rössler J, Bernasconi M. Quantum Dot-Based Screening Identifies F3 Peptide and Reveals Cell Surface Nucleolin as a Therapeutic Target for Rhabdomyosarcoma. Cancers (Basel) 2022; 14:5048. [PMID: 36291832 PMCID: PMC9600270 DOI: 10.3390/cancers14205048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Active drug delivery by tumor-targeting peptides is a promising approach to improve existing therapies for rhabdomyosarcoma (RMS), by increasing the therapeutic effect and decreasing the systemic toxicity, e.g., by drug-loaded peptide-targeted nanoparticles. Here, we tested 20 different tumor-targeting peptides for their ability to bind to two RMS cell lines, Rh30 and RD, using quantum dots Streptavidin and biotin-peptides conjugates as a model for nanoparticles. Four peptides revealed a very strong binding to RMS cells: NCAM-1-targeting NTP peptide, nucleolin-targeting F3 peptide, and two Furin-targeting peptides, TmR and shTmR. F3 peptide showed the strongest binding to all RMS cell lines tested, low binding to normal control myoblasts and fibroblasts, and efficient internalization into RMS cells demonstrated by the cytoplasmic delivery of the Saporin toxin. The expression of the nucleophosphoprotein nucleolin, the target of F3, on the surface of RMS cell lines was validated by competition with the natural ligand lactoferrin, by colocalization with the nucleolin-binding aptamer AS1411, and by the marked sensitivity of RMS cell lines to the growth inhibitory nucleolin-binding N6L pseudopeptide. Taken together, our results indicate that nucleolin-targeting by F3 peptide represents a potential therapeutic approach for RMS.
Collapse
Affiliation(s)
- Dzhangar Dzhumashev
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Andrea Timpanaro
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Safa Ali
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Andrea J. De Micheli
- Department of Oncology, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 3032 Zurich, Switzerland
| | - Kamel Mamchaoui
- Centre de Recherche en Myologie, Institut de Myologie, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Ilaria Cascone
- IMRB, INSERM, University Paris Est Creteil, 94010 Creteil, France
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d’Investigation Clinique Biothérapie, 94010 Créteil, France
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 3032 Zurich, Switzerland
| |
Collapse
|
10
|
Arnau Del Valle C, Hirsch T, Marin M. Recent Advances in Near Infrared Upconverting Nanomaterials for Targeted Photodynamic Therapy of Cancer. Methods Appl Fluoresc 2022; 10. [PMID: 35447614 DOI: 10.1088/2050-6120/ac6937] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/21/2022] [Indexed: 11/11/2022]
Abstract
Photodynamic therapy (PDT) is a well-established treatment of cancer that uses the toxic reactive oxygen species, including singlet oxygen (1O2), generated by photosensitiser drugs following irradiation of a specific wavelength to destroy the cancerous cells and tumours. Visible light is commonly used as the excitation source in PDT, which is not ideal for cancer treatment due to its reduced tissue penetration, and thus inefficiency to treat deep-lying tumours. Additionally, these wavelengths exhibit elevated autofluorescence background from the biological tissues which hinders optical biomedical imaging. An alternative to UV-Vis irradiation is the use of near infrared (NIR) excitation for PDT. This can be achieved using upconverting nanoparticles (UCNPs) functionalised with photosensitiser (PS) drugs where UCNPs can be used as an indirect excitation source for the activation of PS drugs yielding to the production of singlet 1O2 following NIR excitation. The use of nanoparticles for PDT is also beneficial due to their tumour targeting capability, either passively via the enhanced permeability and retention (EPR) effect or actively via stimuli-responsive targeting and ligand-mediated targeting (ie. using recognition units that can bind specific receptors only present or overexpressed on tumour cells). Here, we review recent advances in NIR upconverting nanomaterials for PDT of cancer with a clear distinction between those reported nanoparticles that could potentially target the tumour due to accumulation via the EPR effect (passive targeting) and nanoparticle-based systems that contain targeting agents with the aim of actively target the tumour via a molecular recognition process.
Collapse
Affiliation(s)
- Carla Arnau Del Valle
- University of East Anglia, School of Chemistry, Norwich Research Park, Norwich, NR4 7TJ, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Thomas Hirsch
- University of Regensburg, Institute of Analytical Chemistry, Chemo- and Biosensors, Regensburg, 93040, GERMANY
| | - Maria Marin
- University of East Anglia, School of Chemistry, Norwich Research Park, Norwich, NR4 7TJ, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| |
Collapse
|
11
|
Vène E, Jarnouen K, Ribault C, Vlach M, Verres Y, Bourgeois M, Lepareur N, Cammas-Marion S, Loyer P. Circumsporozoite Protein of Plasmodium berghei- and George Baker Virus A-Derived Peptides Trigger Efficient Cell Internalization of Bioconjugates and Functionalized Poly(ethylene glycol)-b-poly(benzyl malate)-Based Nanoparticles in Human Hepatoma Cells. Pharmaceutics 2022; 14:pharmaceutics14040804. [PMID: 35456637 PMCID: PMC9028075 DOI: 10.3390/pharmaceutics14040804] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 02/04/2023] Open
Abstract
In order to identify the peptides, selected from the literature, that exhibit the strongest tropism towards human hepatoma cells, cell uptake assays were performed using biotinylated synthetic peptides bound to fluorescent streptavidin or engrafted onto nanoparticles (NPs), prepared from biotin-poly(ethylene glycol)-block-poly(benzyl malate) (Biot-PEG-b-PMLABe) via streptavidin bridging. Two peptides, derived from the circumsporozoite protein of Plasmodium berghei- (CPB) and George Baker (GB) Virus A (GBVA10-9), strongly enhanced the endocytosis of both streptavidin conjugates and NPs in hepatoma cells, compared to primary human hepatocytes and non-hepatic cells. Unexpectedly, the uptake of CPB- and GBVA10-9 functionalized PEG-b-PMLABe-based NPs by hepatoma cells involved, at least in part, the peptide binding to apolipoproteins, which would promote NP’s interactions with cell membrane receptors of HDL particles. In addition, CPB and GBVA10-9 peptide–streptavidin conjugates favored the uptake by hepatoma cells over that of the human macrophages, known to strongly internalize nanoparticles by phagocytosis. These two peptides are promising candidate ligands for targeting hepatocellular carcinomas.
Collapse
Affiliation(s)
- Elise Vène
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Pôle Pharmacie, Service Hospitalo-Universitaire de Pharmacie, CHU Rennes, F-35033 Rennes, France
| | - Kathleen Jarnouen
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Catherine Ribault
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Manuel Vlach
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- INRAE, Institut AGRO, PEGASE UMR 1348, F-35590 Saint-Gilles, France
| | - Yann Verres
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Mickaël Bourgeois
- CRCINA, Inserm, CNRS, Université de Nantes, F-44000 Nantes, France;
- ARRONAX Cyclotron, F-44817 Saint Herblain, France
| | - Nicolas Lepareur
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Comprehensive Cancer Center Eugène Marquis, F-35000 Rennes, France
- Correspondence: (N.L.); (S.C.-M.); (P.L.)
| | - Sandrine Cammas-Marion
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Institut des Sciences Chimiques de Rennes (ISCR), Ecole Nationale Supérieure de Chimie de Rennes, CNRS UMR 6226, University of Rennes, F-35042 Rennes, France
- Correspondence: (N.L.); (S.C.-M.); (P.L.)
| | - Pascal Loyer
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Correspondence: (N.L.); (S.C.-M.); (P.L.)
| |
Collapse
|
12
|
Wang L, Chen H, Wang F, Zhang X. The development of peptide-drug conjugates (PDCs) strategies for paclitaxel. Expert Opin Drug Deliv 2022; 19:147-161. [PMID: 35130795 DOI: 10.1080/17425247.2022.2039621] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Paclitaxel is a powerful and effective anti-tumor drug with wide clinical application. However, there are still some limitations, including poor water solubility, low specificity, and susceptibility to drug resistance. The peptide-drug conjugates (PDCs) represent a rising class of therapeutic drugs, which combines small-molecule chemotherapeutic drugs with highly flexible peptides through a cleavable or non-cleavable linker. When this strategy is applied, the therapeutic effects of paclitaxel can be improved. AREAS COVERED In this review, we discuss the application of the PDCs strategy in paclitaxel, including two parts: the tumor targeting peptide-paclitaxel conjugates and the cell penetrating peptide-paclitaxel conjugates. EXPERT OPINION Combining drugs with multifunctional peptides covalently is an effective strategy for delivering paclitaxel to tumors. Depending on different functional peptides, conjugates can increase the water solubility of paclitaxel, tumor permeability of paclitaxel, the accumulation of paclitaxel in tumor tissues, and enhance the antitumor effect of paclitaxel. In addition, due to the change of cell entry mechanism, partial conjugates can restore the therapeutic activity of paclitaxel against resistant tumors. Notably, in order to better translate into the clinical field in the future, more research should be conducted to ensure the safety and effectiveness of peptide-paclitaxel conjugates.
Collapse
Affiliation(s)
- Longkun 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, Jinan 250012, People's Republic of China
| | - Hongyuan Chen
- Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong University, Jinan 250012, People's Republic of 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, Jinan 250012, People's Republic of China
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, People's Republic of China
| |
Collapse
|
13
|
Pryyma A, Matinkhoo K, Bu YJ, Merkens H, Zhang Z, Bénard F, Perrin DM. Synthesis and preliminary evaluation of octreotate conjugates of bioactive synthetic amatoxins for targeting somatostatin receptor (sstr2) expressing cells. RSC Chem Biol 2022; 3:69-78. [PMID: 35128410 PMCID: PMC8729174 DOI: 10.1039/d1cb00036e] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 09/27/2021] [Indexed: 11/28/2022] Open
Abstract
Targeted cancer therapy represents a paradigm-shifting approach that aims to deliver a toxic payload selectively to target-expressing cells thereby sparing normal tissues the off-target effects associated with traditional chemotherapeutics. Since most targeted constructs rely on standard microtubule inhibitors or DNA-reactive molecules as payloads, new toxins that inhibit other intracellular targets are needed to realize the full potential of targeted therapy. Among these new payloads, α-amanitin has gained attraction as a payload in targeted therapy. Here, we conjugate two synthetic amanitins at different sites to demonstrate their utility as payloads in peptide drug conjugates (PDCs). As an exemplary targeting agent, we chose octreotate, a well-studied somatostatin receptor (sstr2) peptide agonist for the conjugation to synthetic amatoxins via three tailor-built linkers. The linker chemistry permitted the evaluation of one non-cleavable and two cleavable self-immolative conjugates. The immolating linkers were chosen to take advantage of either the reducing potential of the intracellular environment or the high levels of lysosomal proteases in tumor cells to trigger toxin release. Cell-based assays on target-positive Ar42J cells revealed target-specific reduction in viability with up to 1000-fold enhancement in bioactivity compared to the untargeted amatoxins. Altogether, this preliminary study enabled the development of a highly modular synthetic platform for the construction of amanitin-based conjugates that can be readily extended to various targeting moieties.
Collapse
Affiliation(s)
- Alla Pryyma
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Kaveh Matinkhoo
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Yong Jia Bu
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Helen Merkens
- Department of Molecular Oncology, BC Cancer Vancouver BC V5Z 1L3 Canada
| | - Zhengxing Zhang
- Department of Molecular Oncology, BC Cancer Vancouver BC V5Z 1L3 Canada
| | - Francois Bénard
- Department of Molecular Oncology, BC Cancer Vancouver BC V5Z 1L3 Canada
| | - David M Perrin
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| |
Collapse
|
14
|
Li S, Pang X, Zhao J, Zhang Q, Shan Y. Evaluating the single-molecule interactions between targeted peptides and the receptors on living cell membrane. NANOSCALE 2021; 13:17318-17324. [PMID: 34642724 DOI: 10.1039/d1nr05547j] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
As potential ligands, targeted peptides have become an important part in the construction of intelligent drug delivery systems (DDSs). The targeting interaction of peptides with receptors is a key point affecting the efficacy of targeted nano-drugs. Herein, three common peptides (HAIYPRH (T7), YHWYGYTPQNVI (GE11), and RGD) that have been widely used in cancer targeted therapy and tumor diagnostics, targeting the corresponding receptors (transferrin receptor (TfR), epidermal growth factor receptor (EGFR), and ανβ3 integrin receptor), were selected as examples to study the targeting interacton on living cell surface at the single-molecule level by using single-molecule force spectroscopy (SMFS) based on atomic force microscopy (AFM). The dissociation activation energy in the absence of an external force (ΔGβ,0) of T7-TfR, GE11-EGFR, and RGD-ανβ3 integrin is evaluated at single-molecule level. Among these three peptide-receptor pairs, the T7-TfR bond is the most stable with a smaller dissociation kinetic rate constant at zero force (Koff), larger kinetic on-rate constant (Kon), and shorter interaction time (τ). Furthermore, T7 can target TfR even more effectively on A549 cell membrane after treatment with drugs. Our methodology can also be applicable to the study of other ligand targeted DDSs.
Collapse
Affiliation(s)
- Siying Li
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
- School of Chemical Engineering, Changchun University of Technology, Changchun 130012, China
| | - Xuelei Pang
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Jing Zhao
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Qingrong Zhang
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Yuping Shan
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| |
Collapse
|
15
|
Ilangala AB, Lechanteur A, Fillet M, Piel G. Therapeutic peptides for chemotherapy: Trends and challenges for advanced delivery systems. Eur J Pharm Biopharm 2021; 167:140-158. [PMID: 34311093 DOI: 10.1016/j.ejpb.2021.07.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/26/2021] [Accepted: 07/16/2021] [Indexed: 01/10/2023]
Abstract
The past decades witnessed an increasing interest in peptides as clinical therapeutics. Rightfully considered as a potential alternative for small molecule therapy, these remarkable pharmaceuticals can be structurally fine-tuned to impact properties such as high target affinity, selectivity, low immunogenicity along with satisfactory tissue penetration. Although physicochemical and pharmacokinetic challenges have mitigated, to some extent, the clinical applications of therapeutic peptides, their potential impact on modern healthcare remains encouraging. According to recent reports, there are more than 400 peptides under clinical trials and 60 were already approved for clinical use. As the demand for efficient and safer therapy became high, especially for cancers, peptides have shown some exciting developments not only due to their potent antiproliferative action but also when used as adjuvant therapies, either to decrease side effects with tumor-targeted therapy or to enhance the activity of anticancer drugs via transbarrier delivery. The first part of the present review gives an insight into challenges related to peptide product development. Both molecular and formulation approaches intended to optimize peptide's pharmaceutical properties are covered, and some of their current issues are highlighted. The second part offers a comprehensive overview of the emerging applications of therapeutic peptides in chemotherapy from bioconjugates to nanovectorized therapeutics.
Collapse
Affiliation(s)
- Ange B Ilangala
- Laboratory for the Analysis of Medicines, CIRM, University of Liège, Avenue Hippocrate 15, 4000 Liège, Belgium; Laboratory of Pharmaceutical Technology and Biopharmacy, Nanomedicine Development, CIRM, University of Liège, Avenue Hippocrate 15, 4000 Liège, Belgium.
| | - Anna Lechanteur
- Laboratory of Pharmaceutical Technology and Biopharmacy, Nanomedicine Development, CIRM, University of Liège, Avenue Hippocrate 15, 4000 Liège, Belgium
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines, CIRM, University of Liège, Avenue Hippocrate 15, 4000 Liège, Belgium
| | - Géraldine Piel
- Laboratory of Pharmaceutical Technology and Biopharmacy, Nanomedicine Development, CIRM, University of Liège, Avenue Hippocrate 15, 4000 Liège, Belgium
| |
Collapse
|
16
|
Watanabe K, Nawachi T, Okutani R, Ohtsuki T. Photocontrolled apoptosis induction using precursor miR-664a and an RNA carrier-conjugated with photosensitizer. Sci Rep 2021; 11:14936. [PMID: 34294789 PMCID: PMC8298592 DOI: 10.1038/s41598-021-94249-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 07/07/2021] [Indexed: 12/15/2022] Open
Abstract
Methods to spatially induce apoptosis are useful for cancer therapy. To control the induction of apoptosis, methods using light, such as photochemical internalization (PCI), have been developed. We hypothesized that photoinduced delivery of microRNAs (miRNAs) that regulate apoptosis could spatially induce apoptosis. In this study, we identified pre-miR-664a as a novel apoptosis-inducing miRNA via mitochondrial apoptotic pathway. Further, we demonstrated the utility of photoinduced cytosolic dispersion of RNA (PCDR), which is an intracellular RNA delivery method based on PCI. Indeed, apoptosis is spatially regulated by pre-miR-664a and PCDR. In addition, we found that apoptosis induced by pre-miR-664a delivered by PCDR was more rapid than that by lipofection. These results suggest that pre-miR-664a is a nucleic acid drug candidate for cancer therapy and PCDR and pre-miR-664a-based strategies have potential therapeutic uses for diseases affecting various cell types.
Collapse
Affiliation(s)
- Kazunori Watanabe
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3-1-1 Tsushimanaka, Okayama, 700-8530, Japan. .,Department of Biomedical Engineering, Faculty of Engineering, Okayama University, 3-1-1 Tsushimanaka, Okayama, 700-8530, Japan.
| | - Tomoko Nawachi
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3-1-1 Tsushimanaka, Okayama, 700-8530, Japan
| | - Ruriko Okutani
- Department of Biomedical Engineering, Faculty of Engineering, Okayama University, 3-1-1 Tsushimanaka, Okayama, 700-8530, Japan
| | - Takashi Ohtsuki
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3-1-1 Tsushimanaka, Okayama, 700-8530, Japan.,Department of Biomedical Engineering, Faculty of Engineering, Okayama University, 3-1-1 Tsushimanaka, Okayama, 700-8530, Japan
| |
Collapse
|
17
|
Zhou J, Li Y, Huang W, Shi W, Qian H. Source and exploration of the peptides used to construct peptide-drug conjugates. Eur J Med Chem 2021; 224:113712. [PMID: 34303870 DOI: 10.1016/j.ejmech.2021.113712] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/12/2021] [Accepted: 07/17/2021] [Indexed: 12/16/2022]
Abstract
Peptide-drug conjugates (PDCs) are a class of novel molecules widely designed and synthesized for delivering payload drugs. The peptide part plays a vital role in the whole molecule, because they determine the ability of the molecules to penetrate the membrane and target to the specific targets. Here, we introduce the source of different kinds of cell-penetrating peptides (CPPs) and cell-targeting peptides (CTPs) that have been used or could be used in constructing PDCs as well as their latest application in delivering drugs. What's more, the approaches of developing CPPs and CTPs and the techniques to discover novel peptides are focused on and summarized in the review. This review aims to help relevant researchers fast understand the research status of peptides in PDCs and carry forward the process of novel peptides discovery.
Collapse
Affiliation(s)
- Jiaqi Zhou
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yuanyuan Li
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Wenlong Huang
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Wei Shi
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Hai Qian
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China.
| |
Collapse
|
18
|
Synthesis of Poly(Malic Acid) Derivatives End-Functionalized with Peptides and Preparation of Biocompatible Nanoparticles to Target Hepatoma Cells. NANOMATERIALS 2021; 11:nano11040958. [PMID: 33918663 PMCID: PMC8070460 DOI: 10.3390/nano11040958] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022]
Abstract
Recently, short synthetic peptides have gained interest as targeting agents in the design of site-specific nanomedicines. In this context, our work aimed at developing new tools for the diagnosis and/or therapy of hepatocellular carcinoma (HCC) by grafting the hepatotropic George Baker (GB) virus A (GBVA10-9) and Plasmodium circumsporozoite protein (CPB)-derived peptides to the biocompatible poly(benzyl malate), PMLABe. We successfully synthesized PMLABe derivatives end-functionalized with peptides GBVA10-9, CPB, and their corresponding scrambled peptides through a thiol/maleimide reaction. The corresponding nanoparticles (NPs), varying by the nature of the peptide (GBVA10-9, CPB, and their scrambled peptides) and the absence or presence of poly(ethylene glycol) were also successfully formulated using nanoprecipitation technique. NPs were further characterized by dynamic light scattering (DLS), electrophoretic light scattering (ELS) and transmission electron microscopy (TEM), highlighting a diameter lower than 150 nm, a negative surface charge, and a more or less spherical shape. Moreover, a fluorescent probe (DiD Oil) has been encapsulated during the nanoprecipitation process. Finally, preliminary in vitro internalisation assays using HepaRG hepatoma cells demonstrated that CPB peptide-functionalized PMLABe NPs were efficiently internalized by endocytosis, and that such nanoobjects may be promising drug delivery systems for the theranostics of HCC.
Collapse
|
19
|
Liu R, Zuo R, Hudalla GA. Harnessing molecular recognition for localized drug delivery. Adv Drug Deliv Rev 2021; 170:238-260. [PMID: 33484737 PMCID: PMC8274479 DOI: 10.1016/j.addr.2021.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/18/2022]
Abstract
A grand challenge in drug delivery is providing the right dose, at the right anatomic location, for the right duration of time to maximize therapeutic efficacy while minimizing off-target toxicity and other deleterious side-effects. Two general modalities are receiving broad attention for localized drug delivery. In the first, referred to as "targeted accumulation", drugs or drug carriers are engineered to have targeting moieties that promote their accumulation at a specific tissue site from circulation. In the second, referred to as "local anchoring", drugs or drug carriers are inserted directly into the tissue site of interest where they persist for a specified duration of time. This review surveys recent advances in harnessing molecular recognition between proteins, peptides, nucleic acids, lipids, and carbohydrates to mediate targeted accumulation and local anchoring of drugs and drug carriers.
Collapse
Affiliation(s)
- Renjie Liu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ran Zuo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
20
|
Drescher S, van Hoogevest P. The Phospholipid Research Center: Current Research in Phospholipids and Their Use in Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12121235. [PMID: 33353254 PMCID: PMC7766331 DOI: 10.3390/pharmaceutics12121235] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022] Open
Abstract
This review summarizes the research on phospholipids and their use for drug delivery related to the Phospholipid Research Center Heidelberg (PRC). The focus is on projects that have been approved by the PRC since 2017 and are currently still ongoing or have recently been completed. The different projects cover all facets of phospholipid research, from basic to applied research, including the use of phospholipids in different administration forms such as liposomes, mixed micelles, emulsions, and extrudates, up to industrial application-oriented research. These projects also include all routes of administration, namely parenteral, oral, and topical. With this review we would like to highlight possible future research directions, including a short introduction into the world of phospholipids.
Collapse
|
21
|
Abasian P, Shakibi S, Maniati MS, Nouri Khorasani S, Khalili S. Targeted delivery, drug release strategies, and toxicity study of polymeric drug nanocarriers. POLYM ADVAN TECHNOL 2020. [DOI: 10.1002/pat.5168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Payam Abasian
- Department of Chemical Engineering Isfahan University of Technology Isfahan Iran
| | - Sepideh Shakibi
- Department of Textile Engineering Amirkabir University of Technology (Tehran Polytechnique) Tehran Iran
| | - Mohammad Saeed Maniati
- Cellular and Molecular Biology Research Center, Health Research Institute Babol University of Medical Sciences Babol Iran
| | | | - Shahla Khalili
- Department of Chemical Engineering Isfahan University of Technology Isfahan Iran
| |
Collapse
|
22
|
Alijaj N, Moutel S, Gouveia ZL, Gray M, Roveri M, Dzhumashev D, Weber F, Meier G, Luciani P, Rössler JK, Schäfer BW, Perez F, Bernasconi M. Novel FGFR4-Targeting Single-Domain Antibodies for Multiple Targeted Therapies against Rhabdomyosarcoma. Cancers (Basel) 2020; 12:cancers12113313. [PMID: 33182650 PMCID: PMC7696840 DOI: 10.3390/cancers12113313] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 01/08/2023] Open
Abstract
Simple Summary Rhabdomyosarcoma (RMS) accounts for more than 50% of all soft tissue sarcomas in childhood and adolescence. Despite progress and intensified multimodality treatment, prognoses are extremely poor with an overall survival rate of approximately 20% in the advanced stage. Therefore, there is an urgent need for targeted treatment options to improve overall survival rates, and to limit long-term side effects. The fibroblast growth factor receptor 4 (FGFR4) is overexpressed in RMS and other tumors as well. The goal of this work was to select FGFR4 specific single-domain antibodies (sdAb) and to develop FGFR4-targeted therapies. We could show that FGFR4-targeted liposomes have the potential to deliver drugs specifically to FGFR4-positive tumor cells and that chimeric antigen receptor T cells built with the selected antibodies can kill specifically FGFR4-expressing RMS cells. Abstract The fibroblast growth factor receptor 4 (FGFR4) is overexpressed in rhabdomyosarcoma (RMS) and represents a promising target for treatments based on specific and efficient antibodies. Despite progress, there is an urgent need for targeted treatment options to improve survival rates, and to limit long-term side effects. From phage display libraries we selected FGFR4-specific single-domain antibodies (sdAb) binding to recombinant FGFR4 and validated them by flow cytometry, surface plasmon resonance, and fluorescence microscopy. The specificity of the selected sdAb was verified on FGFR4-wild type and FGFR4-knock out cells. FGFR4-sdAb were used to decorate vincristine-loaded liposomes and to generate chimeric antigen receptor (CAR) T cells. First, incubation of RMS cells with FGFR4-sdAb revealed that FGFR4-sdAb can block FGF19-FGFR4 signaling via the MAPK pathway and could therefore serve as therapeutics for FGFR4-dependent cancers. Second, FGFR4-targeted vincristine-loaded liposomes bound specifically to RMS cells and were internalized by the receptor, demonstrating the potential for active drug delivery to the tumor. Third, FGFR4-CAR T cells, generated with one sdAb candidate, demonstrated strong and specific cytotoxicity against FGFR4 expressing RMS cells. We selected novel FGFR4-sdAb with high specificity and nano- to picomolar affinities for FGFR4 which have the potential to enable multiple FGFR4-targeted cancer therapy approaches.
Collapse
Affiliation(s)
- Nagjie Alijaj
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Sandrine Moutel
- Institut Curie, PSL Research University, CNRS UMR144, 75248 Paris, France; (S.M.); (Z.L.G.)
- Recombinant Antibody Platform (TAb-IP), Institut Curie, 75248 Paris, France
| | - Zelia L. Gouveia
- Institut Curie, PSL Research University, CNRS UMR144, 75248 Paris, France; (S.M.); (Z.L.G.)
- Honing Biosciences, 75004 Paris, France
| | - Maxim Gray
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
| | - Maurizio Roveri
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
| | - Dzhangar Dzhumashev
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Florian Weber
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland; (F.W.); (P.L.)
| | - Gianmarco Meier
- Institute of Medical Microbiology, University of Zurich, 8006 Zurich, Switzerland;
| | - Paola Luciani
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland; (F.W.); (P.L.)
| | - Jochen K. Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Beat W. Schäfer
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
| | - Franck Perez
- Institut Curie, PSL Research University, CNRS UMR144, 75248 Paris, France; (S.M.); (Z.L.G.)
- Correspondence: (F.P.); (M.B.)
| | - Michele Bernasconi
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Correspondence: (F.P.); (M.B.)
| |
Collapse
|
23
|
Kavand A, Anton N, Vandamme T, Serra CA, Chan-Seng D. Synthesis and functionalization of hyperbranched polymers for targeted drug delivery. J Control Release 2020; 321:285-311. [DOI: 10.1016/j.jconrel.2020.02.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023]
|
24
|
Tripathi VC, Horam S, Singh A, Lata M, Reddy TJ, Arockiaraj J, Pasupuleti M. The discovery of antioxidants in marine microorganisms and their protective effects on the hepatic cells from chemical-induced oxidative stress. Free Radic Res 2020; 54:150-161. [PMID: 32013646 DOI: 10.1080/10715762.2020.1725499] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The marine environment is emerging as a biodiversity resource for the discovery of natural molecules or chemical scaffolds with pharmaceutical activity. Marine microbes have a tremendous ability to sense and respond to their surroundings to survive in a stressful environment by producing different molecules. As oxidative stress is directly or indirectly involved in various pathological conditions in humans, we believe that marine-derived antioxidant molecules will have a great prospect as a novel antioxidant molecule. We, in this work, explored the marine microbial resources from the Gulf of Mannar, Bay of Bengal, India. High-throughput screening of antioxidant molecule producing marine microbes has revealed that extract from Kocuria marina CDMP 10, can effectively reduce the DPPH free radical. Methanolic crude extract obtained by the freeze-thawing was fractionated and purified by using activity guided purification with the help of reverse phase HPLC and analysed through UPLC-MS. Chemical analysis, as well as MS-spectra, indicated that marine bacteria K. marina CDMP 10 derived antioxidant fraction contains the short peptides. The antioxidant activity of the three highly hydrophobic peptides, (Ser-Ser-Gln, Phe-Glu, Asp-Ile and Leu-Glu) was confirmed by in vitro as well as a cell-based assay. These small peptide molecules are noncytotoxic and can prevent the human cells from chemical-induced oxidative stress. Ser-Ser-Gln peptide demonstrated a potent free radical scavenging activity in Hepatocellular carcinoma cell lines. This study suggests that these short peptides from K. marina CDMP 10 may serve as a potential pharmaceutical candidate with antioxidant activity.
Collapse
Affiliation(s)
- Vikash Chandra Tripathi
- Microbiology Division, Council of Scientific and Industrial Research, Central Drug Research Institute, Lucknow, India
| | - Soyar Horam
- Microbiology Division, Council of Scientific and Industrial Research, Central Drug Research Institute, Lucknow, India
| | - Anoop Singh
- Microbiology Division, Council of Scientific and Industrial Research, Central Drug Research Institute, Lucknow, India
| | - Manjul Lata
- Microbiology Division, Council of Scientific and Industrial Research, Central Drug Research Institute, Lucknow, India
| | - Thota Jagadeshwar Reddy
- Council of Scientific and Industrial Research, Indian Institute of Chemical Technology, Hyderabad, India
| | - Jesu Arockiaraj
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, India
| | - Mukesh Pasupuleti
- Microbiology Division, Council of Scientific and Industrial Research, Central Drug Research Institute, Lucknow, India
| |
Collapse
|
25
|
Novel therapeutic interventions in cancer treatment using protein and peptide-based targeted smart systems. Semin Cancer Biol 2019; 69:249-267. [PMID: 31442570 DOI: 10.1016/j.semcancer.2019.08.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 01/12/2023]
Abstract
Cancer, being the most prevalent and resistant disease afflicting any gender, age or social status, is the ultimate challenge for the scientific community. The new generation therapeutics for cancer management has shifted the approach to personalized/precision medicine, making use of patient- and tumor-specific markers for specifying the targeted therapies for each patient. Peptides targeting these cancer-specific signatures hold enormous potential for cancer therapy and diagnosis. The rapid advancements in the combinatorial peptide libraries served as an impetus to the development of multifunctional peptide-based materials for targeted cancer therapy. The present review outlines benefits and shortcomings of peptides as cancer therapeutics and the potential of peptide modified nanomedicines for targeted delivery of anticancer agents.
Collapse
|
26
|
Castillo RR, Lozano D, González B, Manzano M, Izquierdo-Barba I, Vallet-Regí M. Advances in mesoporous silica nanoparticles for targeted stimuli-responsive drug delivery: an update. Expert Opin Drug Deliv 2019; 16:415-439. [PMID: 30897978 PMCID: PMC6667337 DOI: 10.1080/17425247.2019.1598375] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/19/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Mesoporous silica nanoparticles (MSNs) are outstanding nanoplatforms for drug delivery. Herein, the most recent advances to turn MSN-based carriers into minimal side effect drug delivery agents are covered. AREAS COVERED This review summarizes the scientific advances dealing with MSNs for targeted and stimuli-responsive drug delivery since 2015. Delivery aspects to diseased tissues together with approaches to obtain smart MSNs able to respond to internal or external stimuli and their applications are here described. Special emphasis is done on the combination of two or more stimuli on the same nanoplatform and on combined drug therapy. EXPERT OPINION The use of MSNs in nanomedicine is a promising research field because they are outstanding platforms for treating different pathologies. This is possible thanks to their structural, chemical, physical and biological properties. However, there are certain issues that should be overcome to improve the suitability of MSNs for clinical applications. All materials must be properly characterized prior to their in vivo evaluation; furthermore, preclinical in vivo studies need to be standardized to demonstrate the MSNs clinical translation potential.
Collapse
Affiliation(s)
- Rafael R. Castillo
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - Daniel Lozano
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - Blanca González
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - Miguel Manzano
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - Isabel Izquierdo-Barba
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| | - María Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bionorgánica, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)
| |
Collapse
|
27
|
Gurnani P, Sanchez-Cano C, Abraham K, Xandri-Monje H, Cook AB, Hartlieb M, Lévi F, Dallmann R, Perrier S. RAFT Emulsion Polymerization as a Platform to Generate Well-Defined Biocompatible Latex Nanoparticles. Macromol Biosci 2018; 18:e1800213. [PMID: 30085410 DOI: 10.1002/mabi.201800213] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/03/2018] [Indexed: 12/17/2022]
Abstract
Current approaches to generate core-shell nanoparticles for biomedical applications are limited by factors such as synthetic scalability and circulatory desorption of cytotoxic surfactants. Developments in controlled radical polymerization, particularly in dispersed states, represent a promising method of overcoming these challenges. In this work, well-defined PEGylated nanoparticles are synthesized using reversible addition fragmentation chain transfer emulsion polymerization to control particle size and surface composition and were further characterized with light scattering, electron microscopy, and size exclusion chromatography. Importantly, the nanoparticles are found to be tolerated both in vitro and in vivo, without the need for any purification after particle synthesis. Pharmacokinetic and biodistribution studies in mice, following intraperitoneal injection of the nanoparticles, reveal a long (>76 h) circulation time and accumulation in the liver.
Collapse
Affiliation(s)
- Pratik Gurnani
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Carlos Sanchez-Cano
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Kristin Abraham
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Helena Xandri-Monje
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Alexander B Cook
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Matthias Hartlieb
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Francis Lévi
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Robert Dallmann
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Sébastien Perrier
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| |
Collapse
|
28
|
Kirikoshi R, Manabe N, Takahashi O. Phosphate-Catalyzed Succinimide Formation from an NGR-Containing Cyclic Peptide: A Novel Mechanism for Deammoniation of the Tetrahedral Intermediate. Molecules 2018; 23:E2217. [PMID: 30200364 PMCID: PMC6225186 DOI: 10.3390/molecules23092217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/15/2018] [Accepted: 08/30/2018] [Indexed: 01/23/2023] Open
Abstract
Spontaneous deamidation in the Asn-Gly-Arg (NGR) motif that yields an isoAsp-Gly-Arg (isoDGR) sequence has recently attracted considerable attention because of the possibility of application to dual tumor targeting. It is well known that Asn deamidation reactions in peptide chains occur via the five-membered ring succinimide intermediate. Recently, we computationally showed by the B3LYP density functional theory method, that inorganic phosphate and the Arg side chain can catalyze the NGR deamidation using a cyclic peptide, c[CH₂CO⁻NGRC]⁻NH₂. In this previous study, the tetrahedral intermediate of the succinimide formation was assumed to be readily protonated at the nitrogen originating from the Asn side chain by the solvent water before the release of an NH₃ molecule. In the present study, we found a new mechanism for the decomposition of the tetrahedral intermediate that does not require the protonation by an external proton source. The computational method is the same as in the previous study. In the new mechanism, the release of an NH₃ molecule occurs after a proton exchange between the peptide and the phosphate and conformational changes. The rate-determining step of the overall reaction course is the previously reported first step, i.e., the cyclization to form the tetrahedral intermediate.
Collapse
Affiliation(s)
- Ryota Kirikoshi
- Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan.
| | - Noriyoshi Manabe
- Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan.
| | - Ohgi Takahashi
- Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan.
| |
Collapse
|
29
|
Kao C, Chandna R, Ghode A, Dsouza C, Chen M, Larsson A, Lim SH, Wang M, Cao Z, Zhu Y, Anand GS, Ge R. Proapoptotic Cyclic Peptide BC71 Targets Cell-Surface GRP78 and Functions as an Anticancer Therapeutic in Mice. EBioMedicine 2018; 33:22-32. [PMID: 29907328 PMCID: PMC6085501 DOI: 10.1016/j.ebiom.2018.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/25/2018] [Accepted: 06/06/2018] [Indexed: 02/08/2023] Open
Abstract
Glucose regulated protein 78 kDa (GRP78) is a recently emerged target for cancer therapy and a biomarker for cancer prognosis. Overexpression of GRP78 is observed in many types of cancers, with the cell-surface GRP78 being preferentially present in cancer cells and cancer blood vessel endothelial cells. Isthmin (ISM) is a secreted high-affinity proapoptotic protein ligand of cell-surface GRP78 that suppresses angiogenesis and tumor growth in mice. The C-terminal AMOP (adhesion-associated domain in MUC4 and other proteins) domain of ISM is critical in mediating its interaction with human umbilical vein endothelial cells (HUVECs). In this work, we report novel cyclic peptides harboring the RKD motif in the ISM AMOP domain that function as proapoptotic ligands of cell-surface GRP78. The most potent peptide, BC71, binds to GRP78 and converge to tumor in mice. Intravenous administration of BC71 suppressed xenograft tumor growth in mice as a single agent, with significant reduction in tumor angiogenesis and upsurge in apoptosis. Fluorescent-labeled BC71 accumulates in tumor in mice by targeting cell-surface GRP78. We show that BC71 triggers apoptosis via cell-surface GRP78 and activates caspase-8 and p53 signaling pathways in HUVECs. Using amide hydrogen-deuterium exchange mass spectrometry (HDXMS), we identified that BC71 preferentially binds to ATP-bound GRP78 via amino acid residues 244-257 of GRP78. Hence, BC71 serves as a valuable prototype for further development of peptidomimetic anticancer drugs targeting cell-surface GRP78 as well as PET imaging agents for cancer prognosis.
Collapse
Affiliation(s)
- Chieh Kao
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Ritu Chandna
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Abhijeet Ghode
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Charlotte Dsouza
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Mo Chen
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Andreas Larsson
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 639798, Singapore
| | - Siau Hoi Lim
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 639798, Singapore
| | - Minjun Wang
- School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai 201203, China
| | - Zhonglian Cao
- School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai 201203, China
| | - Yizhun Zhu
- School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai 201203, China
| | - Ganesh S Anand
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Ruowen Ge
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore.
| |
Collapse
|
30
|
Vandghanooni S, Eskandani M, Barar J, Omidi Y. Recent advances in aptamer-armed multimodal theranostic nanosystems for imaging and targeted therapy of cancer. Eur J Pharm Sci 2018; 117:301-312. [PMID: 29499349 DOI: 10.1016/j.ejps.2018.02.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 02/24/2018] [Accepted: 02/25/2018] [Indexed: 01/17/2023]
Abstract
The side effects of chemotherapeutics during the course of cancer treatment limit their clinical outcomes. The most important mission of the modern cancer therapy modalities is the delivery of anticancer drugs specifically to the target cells/tissue in order to avoid/reduce any inadvertent non-specific impacts on the healthy normal cells. Nanocarriers decorated with a designated targeting ligand such as aptamers (Aps) and antibodies (Abs) are able to deliver cargo molecules to the target cells/tissue without affecting other neighboring cells, resulting in an improved treatment of cancer. For targeted therapy of cancer, different ligands (e.g., protein, peptide, Abs, Aps and small molecules) have widely been used in the development of different targeting drug delivery systems (DDSs). Of these homing agents, nucleic acid Aps show unique targeting potential with high binding affinity to a variety of biological targets (e.g., genes, peptides, proteins, and even cells and organs). Aps have widely been used as the targeting agent, in large part due to their unique 3D structure, simplicity in synthesis and functionalization, high chemical flexibility, low immunogenicity and toxicity, and cell/tissue penetration capability in some cases. Here, in this review, we provide important insights on Ap-decorated multimodal nanosystems (NSs) and discuss their applications in targeted therapy and imaging of cancer.
Collapse
Affiliation(s)
- Somayeh Vandghanooni
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
31
|
Wu Y, Zhang X, Li H, Deng P, Li H, He T, Rong J, Zhao J, Liu Z. A core/shell stabilized polysaccharide-based nanoparticle with intracellular environment-sensitive drug delivery for breast cancer therapy. J Mater Chem B 2018; 6:6646-6659. [DOI: 10.1039/c8tb00633d] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In this work, we developed a novel core/shell chitosan (Cs)/hyaluronan (HA)-based hybrid nanoparticle, i.e. SNX@Cs-SNX/cHA, with good stability in the bloodstream and intracellular environment-sensitive drug delivery for breast cancer therapy.
Collapse
Affiliation(s)
- Yan Wu
- Department of Materials Science and Engineering
- College of Chemistry and Materials Science
- Jinan University
- Guangzhou 510632
- China
| | - Xinyue Zhang
- Guangzhoujinan Biomedicine Research and Development Center
- Guangdong Provincial Key Laboratory of Bioengineering Medicine
- National Engineering Research Center of Genetic Medicine
- Jinan University
- Guangzhou 510632
| | - Huaqiang Li
- Department of Materials Science and Engineering
- College of Chemistry and Materials Science
- Jinan University
- Guangzhou 510632
- China
| | - Pengfei Deng
- Guangzhoujinan Biomedicine Research and Development Center
- Guangdong Provincial Key Laboratory of Bioengineering Medicine
- National Engineering Research Center of Genetic Medicine
- Jinan University
- Guangzhou 510632
| | - Huiru Li
- Department of Materials Science and Engineering
- College of Chemistry and Materials Science
- Jinan University
- Guangzhou 510632
- China
| | - Tianqi He
- Department of Materials Science and Engineering
- College of Chemistry and Materials Science
- Jinan University
- Guangzhou 510632
- China
| | - Jianhua Rong
- Department of Materials Science and Engineering
- College of Chemistry and Materials Science
- Jinan University
- Guangzhou 510632
- China
| | - Jianhao Zhao
- Department of Materials Science and Engineering
- College of Chemistry and Materials Science
- Jinan University
- Guangzhou 510632
- China
| | - Zhong Liu
- Guangzhoujinan Biomedicine Research and Development Center
- Guangdong Provincial Key Laboratory of Bioengineering Medicine
- National Engineering Research Center of Genetic Medicine
- Jinan University
- Guangzhou 510632
| |
Collapse
|
32
|
Zhao J, Chen H, Tang Y, Chen H, Chen G, Yin Y, Li G. Research progresses on the functional polypeptides in the detection and imaging of breast cancer. J Mater Chem B 2018; 6:2510-2523. [DOI: 10.1039/c7tb02541f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Polypeptides as functional groups continue to garner significant interest in the detection and imaging of breast cancer, working as recognition elements, signal sources, building blocks and therapeutic reagents, etc.
Collapse
Affiliation(s)
- Jing Zhao
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| | - Huinan Chen
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| | - Yingying Tang
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| | - Hong Chen
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| | - Guifang Chen
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| | - Yongmei Yin
- Department of Oncology
- The First Affiliated Hospital of Nanjing Medical University
- Nanjing 210029
- China
| | - Genxi Li
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| |
Collapse
|
33
|
Li C, Chen X, Zhang F, He X, Fang G, Liu J, Wang S. Design of Cyclic Peptide Based Glucose Receptors and Their Application in Glucose Sensing. Anal Chem 2017; 89:10431-10438. [DOI: 10.1021/acs.analchem.7b02430] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Chao Li
- Key Laboratory of Food Nutrition
and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xin Chen
- Key Laboratory of Food Nutrition
and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Fuyuan Zhang
- Key Laboratory of Food Nutrition
and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xingxing He
- Key Laboratory of Food Nutrition
and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Guozhen Fang
- Key Laboratory of Food Nutrition
and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jifeng Liu
- Key Laboratory of Food Nutrition
and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shuo Wang
- Key Laboratory of Food Nutrition
and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| |
Collapse
|
34
|
Bu H, Xu X, Chen J, Cui Y, Wang LQ. Synthesis of a hemoglobin-conjugated triblock copolymer for oxygen carrying and specific recognition of cancer cells. RSC Adv 2017. [DOI: 10.1039/c7ra09747f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Considering that hypoxia causes resistance to anti-cancer therapeutics, we synthesized a hemoglobin-based nanocarrier for oxygen carrying and recognition of cancer cells.
Collapse
Affiliation(s)
- Huixuan Bu
- The Department of Polymer Science and Engineering of Zhejiang University
- Hangzhou
- China
| | - Xin Xu
- The Department of Polymer Science and Engineering of Zhejiang University
- Hangzhou
- China
| | - Jiaming Chen
- The Department of Polymer Science and Engineering of Zhejiang University
- Hangzhou
- China
| | - Yuecheng Cui
- The Department of Polymer Science and Engineering of Zhejiang University
- Hangzhou
- China
| | - Li-Qun Wang
- The Department of Polymer Science and Engineering of Zhejiang University
- Hangzhou
- China
| |
Collapse
|