1
|
Foster T, Lim P, Jones M, Wagle SR, Kovacevic B, Ionescu CM, Wong EYM, Mooranian A, Al-Salami H. Polymer-Based Nanoparticles for Inner Ear Targeted Trans Differentiation Gene Therapy. ChemMedChem 2024; 19:e202400038. [PMID: 38818625 DOI: 10.1002/cmdc.202400038] [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: 01/11/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Hearing loss is a significant disability that often goes under recognised, largely due to poor identification, prevention, and treatment. Steps are being made to amend these pitfalls in the investigation of hearing loss, however, the development of a cure to reverse advanced forms remains distant. This review details some current advances in the treatment of hearing loss, with a particular focus on genetic-based nanotechnology and how it may provide a useful avenue for further research. This review presents a broad background on the pathophysiology of hearing loss and some current interventions. We also highlight some potential genes that may be useful in the amelioration of hearing loss. Pathways of cellular differentiation from stem or supporting cell to functional hair cell are covered in detail, as this mechanism represents a key means of regenerating these cell types. Overall, we believe that polymer-based nanotechnology coupled with novel excipients represents a useful area of further research in the treatment of hearing loss, although further studies in this area are required.
Collapse
Affiliation(s)
- Thomas Foster
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia
- Department of Clinical Biochemistry, Pathwest Laboratory Medicine, Royal Perth Hospital, Perth, 6000, Western Australia, Australia
| | - Patrick Lim
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia
| | - Melissa Jones
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Susbin Raj Wagle
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia
| | - Bozica Kovacevic
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia
| | - Corina Mihaela Ionescu
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia
| | - Elaine Y M Wong
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Armin Mooranian
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia
- School of Pharmacy, University of Otago, Dunedin 9016, Otago, New Zealand
| | - Hani Al-Salami
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102, Western Australia, Australia
- Medical School, The University of Western Australia, Crawley, 6009, Western Australia, Australia
| |
Collapse
|
2
|
Saraiva NM, Alves A, Costa PC, Correia-da-Silva M. Click Chemistry in Polymersome Technology. Pharmaceuticals (Basel) 2024; 17:747. [PMID: 38931414 PMCID: PMC11206349 DOI: 10.3390/ph17060747] [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: 05/16/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Polymersomes, self-assembled nanoparticles composed of amphiphilic block copolymers, have emerged as promising versatile nanovesicles with various applications, such as drug delivery, medical imaging, and diagnostics. The integration of click chemistry reactions, specifically the copper [I]-catalysed azide-alkyne cycloaddition (CuAAC), has greatly expanded the functionalisation and bioconjugation capabilities of polymersomes and new drugs, being this synergistic combination explored in this review. It also provides up-to-date examples of previous incorporations of click-compatible moieties (azide and alkyne functional groups) into polymer building blocks, enabling the "click" attachment of various functional groups and ligands, delving into the diverse range of click reactions that have been reported and employed for polymersome copolymer synthesis and the modification of polymersome surfaces, including ligand conjugation and surface modification. Overall, this review explores the current state-of-the-art of the combinatory usage, in recent years, of polymersomes with the click chemistry reaction, highlighting examples of studies of their synthesis and functionalisation strategies.
Collapse
Affiliation(s)
- Nuno M. Saraiva
- LQOF—Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal;
- CIIMAR—Interdisciplinary Center of Marine and Environmental Research, University of Porto, Terminal dos Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal
| | - Ana Alves
- UCIBIO—Applied Molecular Biosciences Unit, MedTech-Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal; (A.A.); (P.C.C.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Paulo C. Costa
- UCIBIO—Applied Molecular Biosciences Unit, MedTech-Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal; (A.A.); (P.C.C.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Marta Correia-da-Silva
- LQOF—Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal;
- CIIMAR—Interdisciplinary Center of Marine and Environmental Research, University of Porto, Terminal dos Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal
| |
Collapse
|
3
|
Synthesis and evaluation of a novel adapter lipid derivative for preparation of cyclic peptide-modified PEGylated liposomes: Application of cyclic RGD peptide. Eur J Pharm Sci 2022; 176:106239. [PMID: 35714942 DOI: 10.1016/j.ejps.2022.106239] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/15/2022] [Accepted: 06/14/2022] [Indexed: 11/20/2022]
Abstract
Peptide ligand modified nanoparticles can simply prepared by post-insertion method to mix pre-formed nanoparticles with peptide-lipid conjugates in an aqueous solution at an optimal temperature. Therefore, water dispersibility of peptide-lipid conjugates is a very important factor for implementing the post-insertion method. We proposed that highly water dispersible peptide-lipid conjugates can be easily synthesized by separately designing novel adapter lipids with different water dispersibility and reacting them with ligands in a highly efficient manner. Adapter lipids have three critical roles; as spacers of ligand-conjugated lipids for efficient ligand presentation, as structures that form discrete molecular weight distributions, and as providing water dispersibility. In this study, we developed a novel adapter-lipid derivative that enables a variety of cyclic peptide modifications using the click reaction. The integrin αvβ3-targeted cyclic RGDfK (cRGD) peptide was selected as the cyclic peptide ligand. We designed a novel alkyne-tagged lipid with a discrete peptide spacer and bound the cRGD peptide using a click reaction to synthesize a cRGD-conjugated lipid with good water dispersibility for the preparation of cRGD-modified PEGylated liposomes using the post-insertion method. We also revealed that cRGD-modified PEGylated liposomes are efficiently associated with integrin αvβ3-expressing murine colon carcinoma (Colon-26) cells in a modification amount- and peptide sequence-dependent manner, showing high cytotoxicity upon loading with doxorubicin. This novel adapter lipid derivative can be used to synthesize various cyclic peptides by click reactions and will provide useful insights for the future development of cyclic peptide-modified PEGylated liposomes.
Collapse
|
4
|
Villela Zumaya AL, Mincheva R, Raquez JM, Hassouna F. Nanocluster-Based Drug Delivery and Theranostic Systems: Towards Cancer Therapy. Polymers (Basel) 2022; 14:1188. [PMID: 35335518 PMCID: PMC8955999 DOI: 10.3390/polym14061188] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
Over the last decades, the global life expectancy of the population has increased, and so, consequently, has the risk of cancer development. Despite the improvement in cancer therapies (e.g., drug delivery systems (DDS) and theranostics), in many cases recurrence continues to be a challenging issue. In this matter, the development of nanotechnology has led to an array of possibilities for cancer treatment. One of the most promising therapies focuses on the assembly of hierarchical structures in the form of nanoclusters, as this approach involves preparing individual building blocks while avoiding handling toxic chemicals in the presence of biomolecules. This review aims at presenting an overview of the major advances made in developing nanoclusters based on polymeric nanoparticles (PNPs) and/or inorganic NPs. The preparation methods and the features of the NPs used in the construction of the nanoclusters were described. Afterwards, the design, fabrication and properties of the two main classes of nanoclusters, namely noble-metal nanoclusters and hybrid (i.e., hetero) nanoclusters and their mode of action in cancer therapy, were summarized.
Collapse
Affiliation(s)
- Alma Lucia Villela Zumaya
- Faculty of Chemical Engineering, University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic;
| | - Rosica Mincheva
- Laboratory of Polymeric and Composite Materials, University of Mons (UMONS), Place du Parc 20, 7000 Mons, Belgium; (R.M.); (J.-M.R.)
| | - Jean-Marie Raquez
- Laboratory of Polymeric and Composite Materials, University of Mons (UMONS), Place du Parc 20, 7000 Mons, Belgium; (R.M.); (J.-M.R.)
| | - Fatima Hassouna
- Faculty of Chemical Engineering, University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic;
| |
Collapse
|
5
|
Day K, Schneible JD, Young AT, Pozdin VA, Van Den Driessche G, Gaffney LA, Prodromou R, Freytes DO, Fourches D, Daniele M, Menegatti S. Photoinduced reconfiguration to control the protein-binding affinity of azobenzene-cyclized peptides. J Mater Chem B 2021; 8:7413-7427. [PMID: 32661544 DOI: 10.1039/d0tb01189d] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The impact of next-generation biorecognition elements (ligands) will be determined by the ability to remotely control their binding activity for a target biomolecule in complex environments. Compared to conventional mechanisms for regulating binding affinity (pH, ionic strength, or chaotropic agents), light provides higher accuracy and rapidity, and is particularly suited for labile targets. In this study, we demonstrate a general method to develop azobenzene-cyclized peptide ligands with light-controlled affinity for target proteins. Light triggers a cis/trans isomerization of the azobenzene, which results in a major structural rearrangement of the cyclic peptide from a non-binding to a binding configuration. Critical to this goal are the ability to achieve efficient photo-isomerization under low light dosage and the temporal stability of both cis and trans isomers. We demonstrated our method by designing photo-switchable peptides targeting vascular cell adhesion marker 1 (VCAM1), a cell marker implicated in stem cell function. Starting from a known VCAM1-binding linear peptide, an ensemble of azobenzene-cyclized variants with selective light-controlled binding were identified by combining in silico design with experimental characterization via spectroscopy and surface plasmon resonance. Variant cycloAZOB[G-VHAKQHRN-K] featured rapid, light-controlled binding of VCAM1 (KD,trans/KD,cis ∼ 130). Biotin-cycloAZOB[G-VHAKQHRN-K] was utilized to label brain microvascular endothelial cells (BMECs), showing co-localization with anti-VCAM1 antibodies in cis configuration and negligible binding in trans configuration.
Collapse
Affiliation(s)
- Kevin Day
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, North Carolina, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Xiao K, Liu Q, Suby N, Xiao W, Agrawal R, Vu M, Zhang H, Luo Y, Li Y, Lam KS. LHRH-Targeted Redox-Responsive Crosslinked Micelles Impart Selective Drug Delivery and Effective Chemotherapy in Triple-Negative Breast Cancer. Adv Healthc Mater 2021; 10:e2001196. [PMID: 33200571 PMCID: PMC7858235 DOI: 10.1002/adhm.202001196] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/20/2020] [Indexed: 02/05/2023]
Abstract
Systemic chemotherapy is efficacious against triple-negative breast cancer (TNBC), but it is often associated with serious side effects. Here, a luteinizing hormone-releasing hormone (LHRH) receptor-targeted and tumor microenvironment-responsive nanoparticle system to selectively deliver chemotherapeutic drugs to TNBC cells, is reported. This delivery system (termed "LHRH-DCMs") contains poly(ethylene glycol) and dendritic cholic acid as a micellar carrier, reversible intra-micellar disulfide bond as a redox-responsive crosslink, and synthetic high-affinity (D-Lys)-LHRH peptide as a targeting moiety. LHRH-DCMs exhibit high drug loading efficiency, optimal particle size, good colloidal stability, and glutathione-responsive drug release. As expected, LHRH-DCMs are more efficiently internalized into human TNBC cells through receptor-mediated endocytosis, resulting in stronger cytotoxicity against these cancer cells than the non-targeted counterpart when encapsulated with paclitaxel (PTX). Furthermore, near-infrared fluorescence and magnetic resonance imaging demonstrate that LHRH-DCMs facilitate the tumor distribution and penetration of payloads in three different animal models of breast cancer, including cell line-derived xenograft (CDX), patient-derived xenograft (PDX), and transgenic mammary carcinoma. Finally, in vivo therapeutic studies show that PTX-LHRH-DCMs outperform both the corresponding nontargeted PTX-DCMs and the current clinical formulation (Taxol) in an orthotopic TNBC model. These results provide new insights into approaches for precise drug delivery of TNBC.
Collapse
Affiliation(s)
- Kai Xiao
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Qiangqiang Liu
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Nell Suby
- Department of Obstetrics and Gynecology, School of Medicine, University of California, Davis, CA, 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Rinki Agrawal
- Department of Obstetrics and Gynecology, School of Medicine, University of California, Davis, CA, 95817, USA
| | - Michael Vu
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Hongyong Zhang
- Division of Hematology & Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis, CA, 95817, USA
| | - Yan Luo
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Yuanpei Li
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Kit S Lam
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
- Division of Hematology & Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis, CA, 95817, USA
| |
Collapse
|
7
|
Liu X, Zhang Q, Knoll W, Liedberg B, Wang Y. Rational Design of Functional Peptide-Gold Hybrid Nanomaterials for Molecular Interactions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2000866. [PMID: 32743897 DOI: 10.1002/adma.202000866] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/21/2020] [Indexed: 05/12/2023]
Abstract
Gold nanoparticles (AuNPs) have been extensively used for decades in biosensing-related development due to outstanding optical properties. Peptides, as newly realized functional biomolecules, are promising candidates of replacing antibodies, receptors, and substrates for specific molecular interactions. Both peptides and AuNPs are robust and easily synthesized at relatively low cost. Hence, peptide-AuNP-based bio-nano-technological approaches have drawn increasing interest, especially in the field of molecular targeting, cell imaging, drug delivery, and therapy. Many excellent works in these areas have been reported: demonstrating novel ideas, exploring new targets, and facilitating advanced diagnostic and therapeutic technologies. Importantly, some of them also have been employed to address real practical problems, especially in remote and less privileged areas. This contribution focuses on the application of peptide-gold hybrid nanomaterials for various molecular interactions, especially in biosensing/diagnostics and cell targeting/imaging, as well as for the development of highly active antimicrobial/antifouling coating strategies. Rationally designed peptide-gold nanomaterials with functional properties are discussed along with future challenges and opportunities.
Collapse
Affiliation(s)
- Xiaohu Liu
- School of Biomedical Engineering, School of Ophthalmology & Optometry, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Xinsan Road 16, Wenzhou, 325001, China
| | - Qingwen Zhang
- School of Biomedical Engineering, School of Ophthalmology & Optometry, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Xinsan Road 16, Wenzhou, 325001, China
| | - Wolfgang Knoll
- Austrian Institute of Technology, Giefinggasse 4, Vienna, 1210, Austria
| | - Bo Liedberg
- Centre for Biomimetic Sensor Science, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Yi Wang
- School of Biomedical Engineering, School of Ophthalmology & Optometry, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Xinsan Road 16, Wenzhou, 325001, China
| |
Collapse
|
8
|
Rijpkema S, Langens SGHA, van der Kolk MR, Gavriel K, Toebes BJ, Wilson DA. Modular Approach to the Functionalization of Polymersomes. Biomacromolecules 2020; 21:1853-1864. [PMID: 32032491 PMCID: PMC7218747 DOI: 10.1021/acs.biomac.9b01734] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/06/2020] [Indexed: 01/17/2023]
Abstract
Functionalizing polymersomes remains a challenge due to the limitation in reaction conditions applicable to the chemistry on the surface, hindering their application for selective targeting. In order to overcome this limitation, functionalization can be introduced right before the self-assembly. Here, we have synthesized a library (32 examples) of PEG-b-PS and PEG-b-PDLLA with various functional groups derived from the amine-functionalized polymers, leading to functionally active polymersomes. We show that polymersome formation is possible via the general method with all functionalized groups and that these handles are present on the surface and are able to undergo reactions. Additionally, this methodology provides a general synthetic tool to tailor the functional group of the polymersome right before self-assembly, without limitation on the reaction conditions.
Collapse
Affiliation(s)
- Sjoerd
J. Rijpkema
- Institute for Molecules and
Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Sabine G. H. A. Langens
- Institute for Molecules and
Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Marnix R. van der Kolk
- Institute for Molecules and
Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Katerina Gavriel
- Institute for Molecules and
Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - B. Jelle Toebes
- Institute for Molecules and
Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Daniela A. Wilson
- Institute for Molecules and
Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| |
Collapse
|
9
|
Molavipordanjani S, Hosseinimehr SJ. Strategies for Conjugation of Biomolecules to Nanoparticles as Tumor Targeting Agents. Curr Pharm Des 2019; 25:3917-3926. [DOI: 10.2174/1381612825666190903154847] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022]
Abstract
Combination of nanotechnology, biochemistry, chemistry and biotechnology provides the opportunity
to design unique nanoparticles for tumor targeting, drug delivery, medical imaging and biosensing. Nanoparticles
conjugated with biomolecules such as antibodies, peptides, vitamins and aptamer can resolve current challenges
including low accumulation, internalization and retention at the target site in cancer diagnosis and therapy
through active targeting. In this review, we focus on different strategies for conjugation of biomolecules to
nanoparticles such as inorganic nanoparticles (iron oxide, gold, silica and carbon nanoparticles), liposomes, lipid
and polymeric nanoparticles and their application in tumor targeting.
Collapse
Affiliation(s)
- Sajjad Molavipordanjani
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
10
|
Abstract
The identification of markers expressed by pathological cells or their microenvironment would help to distinguish such cells from the normal tissues. The strategies derived from this theory can be a promising modality for imaging and treating diseases. LyP-1, a tumor homing peptide, can selectively bind to its receptor p32 protein overexpressed in various tumor-associated cells and atherosclerotic plaque macrophages. During recent decades, multiple types of LyP-1-based imaging probes and drug delivery systems have been designed and developed for diagnostic and therapeutic applications. This review first introduces LyP-1 and its receptor p32, as well as its homing, internalization and proapoptotic properties. Next, we highlight recent studies focusing on the applications of LyP-1-based strategies in the diagnosis and treatment of tumors, metastatic lesions, and atherosclerotic plaques. Finally, several limitations in the clinical translation of LyP-1-based bioconjugates are summarized.
Collapse
Affiliation(s)
- Ningning Song
- a Department of Nuclear Medicine , Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , People's Republic of China
| | - Lingzhou Zhao
- a Department of Nuclear Medicine , Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , People's Republic of China
| | - Meilin Zhu
- b School of Basic Medical Sciences, Ningxia Medical University , Yinchuan , People's Republic of China
| | - Jinhua Zhao
- a Department of Nuclear Medicine , Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , People's Republic of China
| |
Collapse
|
11
|
Rijpkema SJ, Toebes BJ, Maas MN, Kler NRM, Wilson DA. Designing Molecular Building Blocks for Functional Polymersomes. Isr J Chem 2019. [DOI: 10.1002/ijch.201900039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Sjoerd J. Rijpkema
- Institute for Molecules and MaterialsRadboud University Nijmegen Heyendaalseweg 135 6525 AJ Nijmegen The Netherlands
| | - B. Jelle Toebes
- Institute for Molecules and MaterialsRadboud University Nijmegen Heyendaalseweg 135 6525 AJ Nijmegen The Netherlands
| | - Marijn N. Maas
- Institute for Molecules and MaterialsRadboud University Nijmegen Heyendaalseweg 135 6525 AJ Nijmegen The Netherlands
- Department of Physics, Chemistry and PharmacyUniversity of Southern Denmark Campusvej 55 5230 Odense Denmark
| | - Noël R. M. Kler
- Institute for Molecules and MaterialsRadboud University Nijmegen Heyendaalseweg 135 6525 AJ Nijmegen The Netherlands
| | - Daniela A. Wilson
- Institute for Molecules and MaterialsRadboud University Nijmegen Heyendaalseweg 135 6525 AJ Nijmegen The Netherlands
| |
Collapse
|
12
|
Timur SS, Yöyen-Ermiş D, Esendağlı G, Yonat S, Horzum U, Esendağlı G, Gürsoy RN. Efficacy of a novel LyP-1-containing self-microemulsifying drug delivery system (SMEDDS) for active targeting to breast cancer. Eur J Pharm Biopharm 2019; 136:138-146. [PMID: 30660694 DOI: 10.1016/j.ejpb.2019.01.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/02/2019] [Accepted: 01/16/2019] [Indexed: 12/15/2022]
Abstract
An ideal cancer therapy targets the tumor cells selectively without damaging healthy tissues. Even though the tumor-specific markers are limited, these molecules can be used for the delivery of anti-cancer drugs as an active targeting strategy. Since the lymphatic system plays a critical role in the dissemination of cancer cells, the drugs directed through lymphatics can feasibly reach to the sites of metastasis. LyP-1 is a peptide that binds to the p32 receptor which is highly expressed not only on the lymphatic endothelium but also on the malignant cells; thus, making this peptide ligand a preferable candidate to mediate active targeting of lymphatics and cancer cells. In this study, different formulations of LyP-1 containing lipid-based nanopharmaceutics so-called self-microemulsifying drug delivery systems (SMEDDS) were developed and tested for their efficacy in targeting breast cancer. Following the selection of non-toxic formulation, doxorubicin hydrochloride and LyP-1 were co-administered in the SMEDDS, which resulted in a significant increase in in vitro cytotoxicity in p32-expressing breast cancer cells, 4T1 and MDA-MB-231. Accordingly, the uptake of LyP-1 in the SMEDDS by the cancer cells was demonstrated. The expression of p32 was detected in the 4T1 tumor tissues which were efficiently targeted with LyP-1 in the SMEDDS. When doxorubicin was co-administrated with LyP-1 in SMEDDS via intraperitonial administration, tumor growth and metastasis were significantly reduced. In conclusion, a novel and efficacious SMEDDS formulation containing LyP-1 with a droplet size less than 100 nm was developed for the lymphatic targeting of breast cancer.
Collapse
Affiliation(s)
- Selin S Timur
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Diğdem Yöyen-Ermiş
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Güldal Esendağlı
- Department of Medical Pathology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Selcen Yonat
- Department of Medical Pathology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Utku Horzum
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Güneş Esendağlı
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - R Neslihan Gürsoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
13
|
Large DE, Soucy JR, Hebert J, Auguste DT. Advances in Receptor-Mediated, Tumor-Targeted Drug Delivery. ADVANCED THERAPEUTICS 2019; 2:1800091. [PMID: 38699509 PMCID: PMC11064891 DOI: 10.1002/adtp.201800091] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Indexed: 02/06/2023]
Abstract
Receptor-mediated drug delivery presents an opportunity to enhance therapeutic efficiency by accumulating drug within the tissue of interest and reducing undesired, off-target effects. In cancer, receptor overexpression is a platform for binding and inhibiting pathways that shape biodistribution, toxicity, cell binding and uptake, and therapeutic function. This review will identify tumor-targeted drug delivery vehicles and receptors that show promise for clinical translation based on quantitative in vitro and in vivo data. The authors describe the rationale to engineer a targeted drug delivery vehicle based on the ligand, chemical conjugation method, and type of drug delivery vehicle. Recent advances in multivalent targeting and ligand organization on tumor accumulation are discussed. Revolutionizing receptor-mediated drug delivery may be leveraged in the therapeutic delivery of chemotherapy, gene editing tools, and epigenetic drugs.
Collapse
Affiliation(s)
- Danielle E Large
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| | - Jonathan R Soucy
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| | - Jacob Hebert
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| | - Debra T Auguste
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| |
Collapse
|
14
|
Alhajj N, Chee CF, Wong TW, Rahman NA, Abu Kasim NH, Colombo P. Lung cancer: active therapeutic targeting and inhalational nanoproduct design. Expert Opin Drug Deliv 2018; 15:1223-1247. [DOI: 10.1080/17425247.2018.1547280] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Nasser Alhajj
- Non-Destructive Biomedical and Pharmaceutical Research Centre, iPROMISE, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
| | - Chin Fei Chee
- Nanotechnology & Catalysis Research Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, iPROMISE, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
| | - Noorsaadah Abd Rahman
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- Wellness Research Cluster, Institute of Research Management & Monitoring, University of Malaya, Kuala Lumpur, Malaysia
| | - Paolo Colombo
- Dipartimento di Farmacia, Università degli Studi di Parma, Parma, Italy
| |
Collapse
|
15
|
Cui L, Xiong C, Zhou M, Shi S, Chow DSL, Li C. Integrin αvβ3-Targeted [ 64Cu]CuS Nanoparticles for PET/CT Imaging and Photothermal Ablation Therapy. Bioconjug Chem 2018; 29:4062-4071. [PMID: 30404438 DOI: 10.1021/acs.bioconjchem.8b00690] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Copper sulfide (CuS) nanoparticles have been considered one of the most clinical relevant nanosystems because of their straightforward chemistry, small particle size, low toxicity, and intrinsic theranostic characteristics. In our previous studies, radioactive [64Cu]CuS nanoparticles were successfully developed to be used as efficient radiotracers for positron emission tomography and for photothermal ablation therapy of cancer cells using near-infrared laser irradiation. However, the major challenge of CuS nanoparticles as a theranostic platform is the lack of a means for effective targeted delivery to the tumor site. To overcome this challenge, we designed and synthesized angiogenesis-targeting [64Cu]CuS nanoparticles, which are coupled with cyclic RGDfK peptide [c(RGDfK)] through polyethylene glycol (PEG) linkers using click chemistry. In assessing their tumor-targeting efficacy, we found that the tumor uptakes of [64Cu]CuS-PEG-c(RGDfK) nanoparticles at 24 h after intravenous injection were significantly greater (8.6% ± 1.4% injected dose/gram of tissue) than those of nontargeted [64Cu]CuS-PEG nanoparticles (4.3% ± 1.2% injected dose/gram of tissue, p < 0.05). Irradiation of tumors in mice administered [64Cu]CuS-PEG-c(RGDfK) nanoparticles induced 98.7% necrotic areas. In contrast, irradiation of tumors in mice administered nontargeted CuS-PEG nanoparticles induced 59% necrotic areas ( p < 0.05). The angiogenesis-targeting [64Cu]CuS nanoparticles may serve as a promising platform for image-guided ablation therapy with high efficacy and minimal side effects in future clinical translation of this novel class of multifunctional nanomaterials.
Collapse
Affiliation(s)
- Lili Cui
- Department of Pharmacological and Pharmaceutical Sciences , College of Pharmacy, University of Houston , Houston , Texas 77024 , United States
| | - Chiyi Xiong
- Department of Cancer Systems Imaging , The University of Texas MD Anderson Cancer Center , Houston , Texas 77054-1907 , United States
| | - Min Zhou
- Department of Cancer Systems Imaging , The University of Texas MD Anderson Cancer Center , Houston , Texas 77054-1907 , United States.,Institute of Translational Medicine, School of Medicine , Zhejiang University , Hangzhou , 310029 , China
| | - Sixiang Shi
- Department of Cancer Systems Imaging , The University of Texas MD Anderson Cancer Center , Houston , Texas 77054-1907 , United States
| | - Diana S-L Chow
- Department of Pharmacological and Pharmaceutical Sciences , College of Pharmacy, University of Houston , Houston , Texas 77024 , United States
| | - Chun Li
- Department of Cancer Systems Imaging , The University of Texas MD Anderson Cancer Center , Houston , Texas 77054-1907 , United States
| |
Collapse
|
16
|
Celentano W, Battistella J, Silvestri IP, Bruni R, Huang X, Li M, Messa P, Ordanini S, Cellesi F. Engineered polyester-PEG nanoparticles prepared through a “grafting through” strategy and post-functionalization via Michael type addition. REACT FUNCT POLYM 2018. [DOI: 10.1016/j.reactfunctpolym.2018.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
17
|
Astakhova K, Ray R, Taskova M, Uhd J, Carstens A, Morris K. "Clicking" Gene Therapeutics: A Successful Union of Chemistry and Biomedicine for New Solutions. Mol Pharm 2018; 15:2892-2899. [PMID: 29300491 PMCID: PMC6078818 DOI: 10.1021/acs.molpharmaceut.7b00765] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The use of nucleic acid, DNA and RNA, based strategies to disrupt gene expression as a therapeutic is quickly emerging. Indeed, synthetic oligonucleotides represent a major component of modern gene therapeutics. However, the efficiency and specificity of intracellular uptake for nonmodified oligonucleotides is rather poor. Utilizing RNA based oligonucleotides as therapeutics is even more challenging to deliver, due to extremely fast enzymatic degradation of the RNAs. RNAs get rapidly degraded in vivo and demonstrate large off-target binding events when they can reach and enter the desired target cells. One approach that holds much promise is the utilization of "click chemistry" to conjugate receptor or cell specific targeting molecules directly to the effector oligonucleotides. We discuss here the applications of the breakthrough technology of CuAAC click chemistry and the immense potential in utilizing "click chemistry" in the development of new age targeted oligonucleotide therapeutics.
Collapse
Affiliation(s)
- Kira Astakhova
- Department of Chemistry, Technical University of Denmark, 206 Kemitorvet, 2800 Kgs Lyngby, Denmark
| | - Roslyn Ray
- Center for Gene Therapy, City of Hope – Beckman Research Institute and Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope. 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Maria Taskova
- Department of Chemistry, Technical University of Denmark, 206 Kemitorvet, 2800 Kgs Lyngby, Denmark
| | - Jesper Uhd
- Department of Chemistry, Technical University of Denmark, 206 Kemitorvet, 2800 Kgs Lyngby, Denmark
| | - Annika Carstens
- Department of Chemistry, Technical University of Denmark, 206 Kemitorvet, 2800 Kgs Lyngby, Denmark
| | - Kevin Morris
- Center for Gene Therapy, City of Hope – Beckman Research Institute and Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope. 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| |
Collapse
|
18
|
Gardouh AR, Barakat BM, Qushawy MKE, El-Kazzaz AY, Sami MM, Zaitone SA. Antitumor activity of a molecularly imprinted nanopreparation of 5-flurouracil against Ehrlich's carcinoma solid tumors grown in mice: Comparison to free 5-flurouracil. Chem Biol Interact 2018; 295:52-63. [PMID: 29678497 DOI: 10.1016/j.cbi.2018.04.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/05/2018] [Accepted: 04/16/2018] [Indexed: 11/17/2022]
Abstract
Recently, nanotechnology has received great attention in war against cancer. The present study investigated the antitumor efficacy of molecularly imprinted nanopreparation of 5-fluorouracil (nano-5-FU) against Ehrlich ascites carcinoma (EAC) solid tumors grown in mice. Tumor cells were transplanted into female albino mice. Mice were allocated into 5 groups; Group 1: control EAC bearing mice. Groups 2&3: EAC-bearing mice treated orally with 5-FU (5 and 10 mg/kg) twice weekly. Groups 4&5: EAC bearing mice treated with nano-5-FU (5 and 10 mg/kg) twice weekly. Treatment with nano-5-FU showed higher antitumor effect compared to free 5-FU as indicated by enhanced apoptosis and reduction in tumor weight. Additionally, lower number of mitotic figures and greater area for necrosis were observed in the tumor specimens alongside with a decline in the number of intratumoral proliferating nuclei in comparison to free 5-FU. Furthermore, the results showed a significant down-regulation in tumoral expression of caspase-3 and vascular endothelial growth factor. Together, these results further support the potential of using nanotechnology to enhance anticancer efficacy of 5-FU.
Collapse
Affiliation(s)
- Ahmed R Gardouh
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.
| | - Bassant M Barakat
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt; Department of Clinical Pharmacy, College of Clinical Pharmacy, Al-Baha University, Al-Baha, Saudi Arabia.
| | - Mona K E Qushawy
- Department of Pharmaceutics, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Industries, Sinai University, El-Arish, North Sinai, Egypt.
| | - Amany Y El-Kazzaz
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Manal M Sami
- Department of Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
19
|
Degli Esposti L, Carella F, Adamiano A, Tampieri A, Iafisco M. Calcium phosphate-based nanosystems for advanced targeted nanomedicine. Drug Dev Ind Pharm 2018. [PMID: 29528248 DOI: 10.1080/03639045.2018.1451879] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Synthetic calcium phosphates (CaPs) are the most widely accepted bioceramics for the repair and reconstruction of bone tissue defects. The recent advancements in materials science have prompted a rapid progress in the preparation of CaPs with nanometric dimensions, tailored surface characteristics, and colloidal stability opening new perspectives in their use for applications not strictly related to bone. In particular, the employment of CaPs nanoparticles as carriers of therapeutic and imaging agents has recently raised great interest in nanomedicine. CaPs nanoparticles, as well as other kinds of nanoparticles, can be engineered to specifically target the site of the disease (cells or organs), thus minimizing their dispersion in the body and undesired organism-nanoparticles interactions. The most promising and efficient approach to improve their specificity is the 'active targeting', where nanoparticles are conjugated with a targeting moiety able to recognize and bind with high efficacy and selectivity to receptors that are highly expressed only in the therapeutic site. The aim of this review is to give an overview on advanced targeted nanomedicine with a focus on the most recent reports on CaP nanoparticles-based systems, specifically designed for the active targeting. The distinctive characteristics of CaP nanoparticles with respect to the other kinds of nanomaterials used in nanomedicine are also discussed.
Collapse
Affiliation(s)
- Lorenzo Degli Esposti
- a Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR) , Faenza , Italy
| | - Francesca Carella
- a Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR) , Faenza , Italy
| | - Alessio Adamiano
- a Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR) , Faenza , Italy
| | - Anna Tampieri
- a Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR) , Faenza , Italy
| | - Michele Iafisco
- a Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR) , Faenza , Italy
| |
Collapse
|
20
|
Greene MK, Richards DA, Nogueira JCF, Campbell K, Smyth P, Fernández M, Scott CJ, Chudasama V. Forming next-generation antibody-nanoparticle conjugates through the oriented installation of non-engineered antibody fragments. Chem Sci 2017; 9:79-87. [PMID: 29629076 PMCID: PMC5869316 DOI: 10.1039/c7sc02747h] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/08/2017] [Indexed: 12/15/2022] Open
Abstract
Enabling oriented installation of non-engineered antibody fragments on nanoparticle surfaces to create next-generation antibody–nanoparticle conjugates.
The successful development of targeted nanotherapeutics is contingent upon the conjugation of therapeutic nanoparticles to target-specific ligands, with particular emphasis being placed on antibody-based ligands. Thus, new methods that enable the covalent and precise installation of targeting antibodies to nanoparticle surfaces are greatly desired, especially those which do not rely on costly and time-consuming antibody engineering techniques. Herein we present a novel method for the highly controlled and oriented covalent conjugation of non-engineered antibody F(ab) fragments to PLGA–PEG nanoparticles using disulfide-selective pyridazinedione linkers and strain-promoted alkyne–azide click chemistry. Exemplification of this method with trastuzumab and cetuximab showed significant improvements in both conjugation efficiency and antigen binding capability, when compared to commonly employed strategies for antibody–nanoparticle construction. This new approach paves the way for the development of antibody-targeted nanomedicines with improved paratope availability, reproducibility and uniformity to enhance both biological activity and ease of manufacture.
Collapse
Affiliation(s)
- Michelle K Greene
- Centre for Cancer Research and Cell Biology , School of Medicine , Dentistry and Biomedical Sciences , Queen's University Belfast , Belfast , UK .
| | | | | | - Katrina Campbell
- Institute for Global Food Security , School of Biological Sciences , Queen's University Belfast , Belfast , UK
| | - Peter Smyth
- Centre for Cancer Research and Cell Biology , School of Medicine , Dentistry and Biomedical Sciences , Queen's University Belfast , Belfast , UK .
| | - Marcos Fernández
- Department of Chemistry , University College London , London , UK .
| | - Christopher J Scott
- Centre for Cancer Research and Cell Biology , School of Medicine , Dentistry and Biomedical Sciences , Queen's University Belfast , Belfast , UK .
| | - Vijay Chudasama
- Department of Chemistry , University College London , London , UK . .,Research Institute for Medicines (iMed.ULisboa) , Faculty of Pharmacy , Universidade de Lisboa , Lisbon , Portugal
| |
Collapse
|
21
|
Olejniczak J, Collet G, Nguyen Huu VA, Chan M, Lee S, Almutairi A. Biorthogonal click chemistry on poly(lactic-co-glycolic acid)-polymeric particles. Biomater Sci 2017; 5:211-215. [DOI: 10.1039/c6bm00721j] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Biodegradable polymeric materials are a key area of investigation in drug delivery and disease treatment.
Collapse
Affiliation(s)
- Jason Olejniczak
- Skaggs School of Pharmacy and Pharmaceutical Sciences
- University of California
- La Jolla
- USA
| | - Guillaume Collet
- Skaggs School of Pharmacy and Pharmaceutical Sciences
- University of California
- La Jolla
- USA
| | - Viet Anh Nguyen Huu
- Skaggs School of Pharmacy and Pharmaceutical Sciences
- University of California
- La Jolla
- USA
| | - Minnie Chan
- Skaggs School of Pharmacy and Pharmaceutical Sciences
- University of California
- La Jolla
- USA
| | - Sangeun Lee
- Skaggs School of Pharmacy and Pharmaceutical Sciences
- University of California
- La Jolla
- USA
| | - Adah Almutairi
- Skaggs School of Pharmacy and Pharmaceutical Sciences
- University of California
- La Jolla
- USA
| |
Collapse
|
22
|
Magnetic mesoporous nanospheres anchored with LyP-1 as an efficient pancreatic cancer probe. Biomaterials 2016; 115:9-18. [PMID: 27871003 DOI: 10.1016/j.biomaterials.2016.11.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 10/10/2016] [Accepted: 11/07/2016] [Indexed: 01/17/2023]
Abstract
Immobilization of a ligand that selectively interacts with cancer cells to nanomaterials can enhance their diagnostic and therapeutic efficiency. In this study, we firstly demonstrate the high expression of receptor for cyclic nine-amino acid peptide LyP-1 (Cys-Gly-Asn-Lys-Arg-Thr-Arg-Gly-Cys) in both mouse and human pancreatic cancer. Based on these findings, sub-50 nm multifunctional superparamagnetic mesoporous nanospheres with surface modified with LyP-1 are rationally designed. Theses nanospheres have a core of silica-protected magnetite nanoparticle and a shell of FITC-labeled mesoporous silica, and they are able to specifically recognize and conjugate with the pancreatic cancer cell in vitro, as verified by the combined techniques of fluorescent imaging and T2 weight magnetic resonance imaging. After systematic administration, these LyP-1 immobilized nanospheres are found to actively target to mouse orthotopic xenograft of pancreatic cancer, which opens up the door for applications in early probing and diagnosis of pancreatic cancer by the multimodal imaging.
Collapse
|
23
|
Clayton KN, Salameh JW, Wereley ST, Kinzer-Ursem TL. Physical characterization of nanoparticle size and surface modification using particle scattering diffusometry. BIOMICROFLUIDICS 2016; 10:054107. [PMID: 27703593 PMCID: PMC5035303 DOI: 10.1063/1.4962992] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/06/2016] [Indexed: 05/23/2023]
Abstract
As the field of colloidal science continues to expand, tools for rapid and accurate physiochemical characterization of colloidal particles will become increasingly important. Here, we present Particle Scattering Diffusometry (PSD), a method that utilizes dark field microscopy and the principles of particle image velocimetry to measure the diffusivity of particles undergoing Brownian motion. PSD measures the diffusion coefficient of particles as small as 30 nm in diameter and is used to characterize changes in particle size and distribution as a function of small, label-free, surface modifications of particles. We demonstrate the rapid sizing of particles using three orders-of-magnitude less sample volume than current standard techniques and use PSD to quantify particle uniformity. Furthermore, PSD is sensitive enough to detect biomolecular surface modifications of nanometer thickness. With these capabilities, PSD can reliably aid in a wide variety of applications, including colloid sizing, particle corona characterization, protein footprinting, and quantifying biomolecule activity.
Collapse
Affiliation(s)
| | - Janelle W Salameh
- Weldon School of Biomedical Engineering, Purdue University , West Lafayette, Indiana 47907, USA
| | | | - Tamara L Kinzer-Ursem
- Weldon School of Biomedical Engineering, Purdue University , West Lafayette, Indiana 47907, USA
| |
Collapse
|
24
|
Wang H, Tang L, Liu Y, Dobrucki IT, Dobrucki LW, Yin L, Cheng J. In Vivo Targeting of Metabolically Labeled Cancers with Ultra-Small Silica Nanoconjugates. Am J Cancer Res 2016; 6:1467-76. [PMID: 27375793 PMCID: PMC4924513 DOI: 10.7150/thno.16003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 05/20/2016] [Indexed: 11/17/2022] Open
Abstract
Unnatural sugar-mediated metabolic labeling of cancer cells, coupled with efficient Click chemistry, has shown great potential for in vivo imaging and cancer targeting. Thus far, chemical labeling of cancer cells has been limited to the small-sized azido groups, with the large-sized and highly hydrophobic dibenzocyclooctyne (DBCO) being correspondingly used as the targeting ligand. However, surface modification of nanomedicines with DBCO groups often suffers from low ligand density, difficult functionalization, and impaired physiochemical properties. Here we report the development of DBCO-bearing unnatural sugars that could directly label LS174T colon cancer cells with DBCO groups and subsequently mediate cancer-targeted delivery of azido-modified silica nanoconjugates with easy functionalization and high azido density in vitro and in vivo. This study, for the first time, demonstrates the feasibility of metabolic labeling of cancer cells with large-sized DBCO groups for subsequent, efficient targeting of azido-modified nanomedicines.
Collapse
|
25
|
Huang J, Li Y, Orza A, Lu Q, Guo P, Wang L, Yang L, Mao H. Magnetic Nanoparticle Facilitated Drug Delivery for Cancer Therapy with Targeted and Image-Guided Approaches. ADVANCED FUNCTIONAL MATERIALS 2016; 26:3818-3836. [PMID: 27790080 PMCID: PMC5077153 DOI: 10.1002/adfm.201504185] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
With rapid advances in nanomedicine, magnetic nanoparticles (MNPs) have emerged as a promising theranostic tool in biomedical applications, including diagnostic imaging, drug delivery and novel therapeutics. Significant preclinical and clinical research has explored their functionalization, targeted delivery, controllable drug release and image-guided capabilities. To further develop MNPs for theranostic applications and clinical translation in the future, we attempt to provide an overview of the recent advances in the development and application of MNPs for drug delivery, specifically focusing on the topics concerning the importance of biomarker targeting for personalized therapy and the unique magnetic and contrast-enhancing properties of theranostic MNPs that enable image-guided delivery. The common strategies and considerations to produce theranostic MNPs and incorporate payload drugs into MNP carriers are described. The notable examples are presented to demonstrate the advantages of MNPs in specific targeting and delivering under image guidance. Furthermore, current understanding of delivery mechanisms and challenges to achieve efficient therapeutic efficacy or diagnostic capability using MNP-based nanomedicine are discussed.
Collapse
Affiliation(s)
- Jing Huang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yuancheng Li
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anamaria Orza
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Peng Guo
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031, USA. Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Liya Wang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
26
|
Hiltebrandt K, Elies K, D'hooge DR, Blinco JP, Barner-Kowollik C. A Light-Activated Reaction Manifold. J Am Chem Soc 2016; 138:7048-54. [PMID: 27151599 DOI: 10.1021/jacs.6b01805] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We introduce an efficient reaction manifold where the rate of a thermally induced ligation can be controlled by a photonic field via two competing reaction channels. The effectiveness of the reaction manifold is evidenced by following the transformations of macromolecular chain termini via high-resolution mass spectrometry and subsequently by selective block copolymer formation. The light-controlled reaction manifold consists of a so-called o-quinodimethane species, a photocaged diene, that reacts in the presence of light with suitable enes in a Diels-Alder reaction and undergoes a transformation into imines with amines in the absence of light. The chemical selectivity of the manifold is controlled by the amount of ene present in the reaction and can be adjusted from 100% imine formation (0% photo product) to 5% imine formation (95% photo product). The reported light-controlled reaction manifold is highly attractive because a simple external field is used to switch the selectivity of specific reaction channels.
Collapse
Affiliation(s)
- Kai Hiltebrandt
- Preparative Macromolecular Chemistry, Institut für Technische Chemie und Polymerchemie, Karlsruhe Institute of Technology (KIT) , Engesserstraße 18, 76128 Karlsruhe, Germany.,Institut für Biologische Grenzflächen (IBG), Karlsruhe Institute of Technology (KIT) , Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Katharina Elies
- Preparative Macromolecular Chemistry, Institut für Technische Chemie und Polymerchemie, Karlsruhe Institute of Technology (KIT) , Engesserstraße 18, 76128 Karlsruhe, Germany
| | - Dagmar R D'hooge
- Department of Chemical Engineering and Technical Chemistry, Laboratory for Chemical Technology, Ghent University , Technologiepark 914, B-9052 Gent, Belgium.,Department of Textiles, Ghent University , Technologiepark 907, B-9052 Gent, Belgium
| | - James P Blinco
- School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology (QUT) , 2 George Street, Brisbane, QLD 4000, Australia
| | - Christopher Barner-Kowollik
- Preparative Macromolecular Chemistry, Institut für Technische Chemie und Polymerchemie, Karlsruhe Institute of Technology (KIT) , Engesserstraße 18, 76128 Karlsruhe, Germany.,Institut für Biologische Grenzflächen (IBG), Karlsruhe Institute of Technology (KIT) , Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany.,School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology (QUT) , 2 George Street, Brisbane, QLD 4000, Australia
| |
Collapse
|
27
|
Chen Y, Xianyu Y, Wu J, Yin B, Jiang X. Click Chemistry-Mediated Nanosensors for Biochemical Assays. Theranostics 2016; 6:969-85. [PMID: 27217831 PMCID: PMC4876622 DOI: 10.7150/thno.14856] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 02/11/2016] [Indexed: 12/19/2022] Open
Abstract
Click chemistry combined with functional nanoparticles have drawn increasing attention in biochemical assays because they are promising in developing biosensors with effective signal transformation/amplification and straightforward signal readout for clinical diagnostic assays. In this review, we focus on the latest advances of biochemical assays based on Cu (I)-catalyzed 1, 3-dipolar cycloaddition of azides and alkynes (CuAAC)-mediated nanosensors, as well as the functionalization of nanoprobes based on click chemistry. Nanoprobes including gold nanoparticles, quantum dots, magnetic nanoparticles and carbon nanomaterials are covered. We discuss the advantages of click chemistry-mediated nanosensors for biochemical assays, and give perspectives on the development of click chemistry-mediated approaches for clinical diagnosis and other biomedical applications.
Collapse
Affiliation(s)
| | | | | | | | - Xingyu Jiang
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing 100190, China
| |
Collapse
|
28
|
Singh M, Bhatnagar P, Mishra S, Kumar P, Shukla Y, Gupta KC. PLGA-encapsulated tea polyphenols enhance the chemotherapeutic efficacy of cisplatin against human cancer cells and mice bearing Ehrlich ascites carcinoma. Int J Nanomedicine 2015; 10:6789-809. [PMID: 26586942 PMCID: PMC4636172 DOI: 10.2147/ijn.s79489] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The clinical success of the applicability of tea polyphenols awaits efficient systemic delivery and bioavailability. Herein, following the concept of nanochemoprevention, which uses nanotechnology for enhancing the efficacy of chemotherapeutic drugs, we employed tea polyphenols, namely theaflavin (TF) and epigallocatechin-3-gallate (EGCG) encapsulated in a biodegradable nanoparticulate formulation based on poly(lactide-co-glycolide) (PLGA) with approximately 26% and 18% encapsulation efficiency, respectively. It was observed that TF/EGCG encapsulated PLGA nanoparticles (NPs) offered an up to ~7-fold dose advantage when compared with bulk TF/EGCG in terms of exerting its antiproliferative effects and also enhanced the anticancer potential of cisplatin (CDDP) in A549 (lung carcinoma), HeLa (cervical carcinoma), and THP-1 (acute monocytic leukemia) cells. Cell cycle analysis revealed that TF/EGCG-NPs were more efficient than bulk TF/EGCG in sensitizing A549 cells to CDDP-induced apoptosis, with a dose advantage of up to 20-fold. Further, TF/EGCG-NPs, alone or in combination with CDDP, were more effective in inhibiting NF-κB activation and in suppressing the expression of cyclin D1, matrix metalloproteinase-9, and vascular endothelial growth factor, involved in cell proliferation, metastasis, and angiogenesis, respectively. EGCG and TF-NPs were also found to be more effective than bulk TF/EGCG in inducing the cleavage of caspase-3 and caspase-9 and Bax/Bcl2 ratio in favor of apoptosis. Further, in vivo evaluation of these NPs in combination with CDDP showed an increase in life span (P<0.05) in mice bearing Ehrlich's ascites carcinoma cells, with apparent regression of tumor volume in comparison with mice treated with bulk doses with CDDP. These results indicate that EGCG and TF-NPs have superior cancer chemosensitization activity when compared with bulk TF/EGCG.
Collapse
Affiliation(s)
- Madhulika Singh
- CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Priyanka Bhatnagar
- CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Delhi, India
| | - Sanjay Mishra
- CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Pradeep Kumar
- CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Delhi, India
| | | | - Kailash Chand Gupta
- CSIR-Indian Institute of Toxicology Research, Lucknow, India ; CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Delhi, India
| |
Collapse
|
29
|
Stolzer L, Vigovskaya A, Barner-Kowollik C, Fruk L. A Self-Reporting Tetrazole-Based Linker for the Biofunctionalization of Gold Nanorods. Chemistry 2015; 21:14309-13. [DOI: 10.1002/chem.201502070] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 11/11/2022]
|
30
|
Elevated expression of HABP1 is a novel prognostic indicator in triple-negative breast cancers. Tumour Biol 2015; 36:4793-9. [DOI: 10.1007/s13277-015-3131-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 01/19/2015] [Indexed: 12/21/2022] Open
|
31
|
Slegerova J, Hajek M, Rehor I, Sedlak F, Stursa J, Hruby M, Cigler P. Designing the nanobiointerface of fluorescent nanodiamonds: highly selective targeting of glioma cancer cells. NANOSCALE 2015; 7:415-20. [PMID: 25132312 DOI: 10.1039/c4nr02776k] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Core-shell nanoparticles based on fluorescent nanodiamonds coated with a biocompatible N-(2-hydroxypropyl)methacrylamide copolymer shell were developed for background-free near-infrared imaging of cancer cells. The particles showed excellent colloidal stability in buffers and culture media. After conjugation with a cyclic RGD peptide they selectively targeted integrin αvβ3 receptors on glioblastoma cells with high internalization efficacy.
Collapse
Affiliation(s)
- Jitka Slegerova
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Flemingovo nam. 2, 166 10, Prague 6, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
32
|
Qin M, Zong H, Kopelman R. Click conjugation of peptide to hydrogel nanoparticles for tumor-targeted drug delivery. Biomacromolecules 2014; 15:3728-34. [PMID: 25162488 DOI: 10.1021/bm501028c] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Here we introduce a modified peptide-decorated polymeric nanoparticle (NP) for cancer cell targeting, which can deliver drugs, such as doxorubicin (Dox), to several kinds of cancer cells. Specifically, we employ a nucleolin-targeting NP, with a matrix based on a copolymer of acrylamide (AAm) and 2-carboxyethyl acrylate (CEA). The negatively charged co(CEA-AAm) NP was conjugated with a nucleolin-targeting F3 peptide using a highly efficient and specific copper(I) catalyzed azide-alkyne click reaction. F3 peptide binds to angiogenic tumor vasculatures and other nucleolin overexpressing tumor cells. Attaching F3 peptide onto the NP increases the NP uptake by the nucleolin-expressing glioma cell line 9L and the breast cancer cell line MCF-7. Notably, the F3-conjugated NPs show much higher uptake by the nucleolin-overexpressing glioma cell line 9L than that by the breast cancer cell line MCF-7, the latter having a lower expression of nucleolin on its plasma membrane surface. Moreover, the F3 peptide also dramatically enhances the uptake of co(CEA-AAm) NPs by the drug-resistant cell line NCI/ADR-RES. Also, with this F3-conjugated co(CEA-AAm) NP, a high loading and slow release of doxorubicin were achieved.
Collapse
Affiliation(s)
- Ming Qin
- Department of Chemistry and ‡Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan , Ann Arbor, Michigan 48109, United States
| | | | | |
Collapse
|
33
|
Breger JC, Fisher B, Samy R, Pollack S, Wang NS, Isayeva I. Synthesis of “click” alginate hydrogel capsules and comparison of their stability, water swelling, and diffusion properties with that of Ca+2crosslinked alginate capsules. J Biomed Mater Res B Appl Biomater 2014; 103:1120-32. [DOI: 10.1002/jbm.b.33282] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 08/08/2014] [Accepted: 09/01/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Joyce C. Breger
- Center for Devices and Radiological Health/Office of Science and Engineering Laboratories/FDA; Silver Spring Maryland 20993-002
- Department of Chemical and Biomolecular Engineering; University of Maryland; Maryland 20742-2115
| | - Benjamin Fisher
- Center for Devices and Radiological Health/Office of Science and Engineering Laboratories/FDA; Silver Spring Maryland 20993-002
| | - Raghu Samy
- Center for Devices and Radiological Health/Office of Science and Engineering Laboratories/FDA; Silver Spring Maryland 20993-002
| | - Steven Pollack
- Center for Devices and Radiological Health/Office of Science and Engineering Laboratories/FDA; Silver Spring Maryland 20993-002
| | - Nam Sun Wang
- Department of Chemical and Biomolecular Engineering; University of Maryland; Maryland 20742-2115
| | - Irada Isayeva
- Center for Drug Evaluation and Research/Office of Pharmaceutical Science/Office of Generic Drugs/Division of Chemistry III/FDA; Silver Spring Maryland 20993-002
| |
Collapse
|
34
|
Lin KY, Kwon EJ, Lo JH, Bhatia SN. Drug-induced amplification of nanoparticle targeting to tumors. NANO TODAY 2014; 9:550-559. [PMID: 29731806 PMCID: PMC5935498 DOI: 10.1016/j.nantod.2014.09.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nanomedicines have the potential to significantly impact cancer therapy by improving drug efficacy and decreasing off-target effects, yet our ability to efficiently home nanoparticles to disease sites remains limited. One frequently overlooked constraint of current active targeting schemes is the relative dearth of targetable antigens within tumors, which restricts the amount of cargo that can be delivered in a tumor-specific manner. To address this limitation, we exploit tumor-specific responses to drugs to construct a cooperative targeting system where a small molecule therapeutic modulates the disease microenvironment to amplify nanoparticle recruitment in vivo. We first administer a vascular disrupting agent, ombrabulin, which selectively affects tumors and leads to locally elevated presentation of the stress-related protein, p32. This increase in p32 levels provides more binding sites for circulating p32-targeted nanoparticles, enhancing their delivery of diagnostic or therapeutic cargos to tumors. We show that this cooperative targeting system recruits over five times higher doses of nanoparticles to tumors and decreases tumor burden when compared with non-cooperative controls. These results suggest that using nanomedicine in conjunction with drugs that enhance the presentation of target antigens in the tumor environment may be an effective strategy for improving the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Kevin Y Lin
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Ester J Kwon
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Justin H Lo
- Medical Scientist Training Program, Harvard Medical School, Boston, MA 02115
- Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | - Sangeeta N Bhatia
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115
- Electrical Engineering and Computer Science, David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| |
Collapse
|
35
|
Martin SE, Ganguly T, Munske GR, Fulton MD, Hopkins MR, Berkman CE, Black ME. Development of inhibitor-directed enzyme prodrug therapy (IDEPT) for prostate cancer. Bioconjug Chem 2014; 25:1752-60. [PMID: 25157916 PMCID: PMC4198102 DOI: 10.1021/bc500362n] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
![]()
Prostate
cancer (PCa) is the second most common cause of cancer
death among American men after lung cancer. Unfortunately, current
therapies do not provide effective treatments for patients with advanced,
metastatic, or hormone refractory disease. Therefore, we seek to generate
therapeutic agents for a novel PCa treatment strategy by delivering
a suicide enzyme (yCDtriple) to a cell membrane bound biomarker
found on PCa cells (prostate-specific membrane antigen (PSMA)). This
approach has resulted in a new PCa treatment strategy reported here
as inhibitor-directed enzyme prodrug therapy (IDEPT). The therapeutic
agents described were generated using a click chemistry reaction between
the unnatural amino acid (p-azidophenylalanine (pAzF)) incorporated into yCDtriple and the dibenzylcyclooctyne
moiety of our PSMA targeting agent (DBCO-PEG4-AH2-TG97). After characterization of the therapeutic agents, we demonstrate
significant PCa cell killing of PSMA-positive cells. Importantly,
we demonstrate that this click chemistry approach can be used to efficiently
couple a therapeutic protein to a targeting agent and may be applicable
to the ablation of other types of cancers and/or malignancies.
Collapse
Affiliation(s)
- Stacy E Martin
- School of Molecular Biosciences and §Department of Chemistry, Washington State University , Pullman, Washington 99164-7520, United States
| | | | | | | | | | | | | |
Collapse
|
36
|
Conde J, Dias JT, Grazú V, Moros M, Baptista PV, de la Fuente JM. Revisiting 30 years of biofunctionalization and surface chemistry of inorganic nanoparticles for nanomedicine. Front Chem 2014; 2:48. [PMID: 25077142 PMCID: PMC4097105 DOI: 10.3389/fchem.2014.00048] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 06/24/2014] [Indexed: 01/04/2023] Open
Abstract
In the last 30 years we have assisted to a massive advance of nanomaterials in material science. Nanomaterials and structures, in addition to their small size, have properties that differ from those of larger bulk materials, making them ideal for a host of novel applications. The spread of nanotechnology in the last years has been due to the improvement of synthesis and characterization methods on the nanoscale, a field rich in new physical phenomena and synthetic opportunities. In fact, the development of functional nanoparticles has progressed exponentially over the past two decades. This work aims to extensively review 30 years of different strategies of surface modification and functionalization of noble metal (gold) nanoparticles, magnetic nanocrystals and semiconductor nanoparticles, such as quantum dots. The aim of this review is not only to provide in-depth insights into the different biofunctionalization and characterization methods, but also to give an overview of possibilities and limitations of the available nanoparticles.
Collapse
Affiliation(s)
- João Conde
- Harvard-MIT Division for Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of TechnologyCambridge, MA, USA
| | - Jorge T. Dias
- Nanotherapy and Nanodiagnostics Group, Instituto de Nanociencia de Aragon, Universidad de ZaragozaZaragoza, Spain
| | - Valeria Grazú
- Nanotherapy and Nanodiagnostics Group, Instituto de Nanociencia de Aragon, Universidad de ZaragozaZaragoza, Spain
| | - Maria Moros
- Nanotherapy and Nanodiagnostics Group, Instituto de Nanociencia de Aragon, Universidad de ZaragozaZaragoza, Spain
| | - Pedro V. Baptista
- CIGMH, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de LisboaCaparica, Portugal
| | - Jesus M. de la Fuente
- Nanotherapy and Nanodiagnostics Group, Instituto de Nanociencia de Aragon, Universidad de ZaragozaZaragoza, Spain
- Fundacion ARAIDZaragoza, Spain
- Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Department of Bio-Nano Science and Engineering, Institute of Nano Biomedicine and Engineering, Research Institute of Translation Medicine, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|
37
|
Dellinger A, Zhou Z, Connor J, Madhankumar AB, Pamujula S, Sayes CM, Kepley CL. Application of fullerenes in nanomedicine: an update. Nanomedicine (Lond) 2014; 8:1191-208. [PMID: 23837857 DOI: 10.2217/nnm.13.99] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Fullerenes are carbon spheres presently being pursued globally for a wide range of applications in nanomedicine. These molecules have unique electronic properties that make them attractive candidates for diagnostic, therapeutic and theranostic applications. Herein, the latest research is discussed on developing fullerene-based therapeutics as antioxidants for inflammatory diseases, their potential as antiviral/bacterial agents, utility as a drug delivery device and the promise of endohedral fullerenes as new MRI contrast agents. The recent discovery that certain fullerene derivatives can stabilize immune effector cells to prevent or inhibit the release of proinflammatory mediators makes them potential candidates for several diseases such as asthma, arthritis and multiple sclerosis. Gadolinium-containing endohedral fullerenes are being pursued as diagnostic MRI contrast agents for several diseases. Finally, a new class of fullerene-based theranostics has been developed, which combine therapeutic and diagnostic capabilities to specifically detect and kill cancer cells.
Collapse
Affiliation(s)
- Anthony Dellinger
- Joint School of Nanoscience & Nanoengineering, 2907 East Lee Street, Greensboro, NC 27401, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Affiliation(s)
- Bethany Powell Gray
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| | - Kathlynn C. Brown
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| |
Collapse
|
39
|
Tang W, Becker ML. “Click” reactions: a versatile toolbox for the synthesis of peptide-conjugates. Chem Soc Rev 2014; 43:7013-39. [DOI: 10.1039/c4cs00139g] [Citation(s) in RCA: 271] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Peptides that comprise the functional subunits of proteins have been conjugated to versatile materials (biomolecules, polymers, surfaces and nanoparticles) in an effort to modulate cell responses, specific binding affinity and/or self-assembly behavior.
Collapse
Affiliation(s)
- Wen Tang
- Department of Polymer Science
- The University of Akron
- Akron, USA
| | - Matthew L. Becker
- Department of Polymer Science
- The University of Akron
- Akron, USA
- Department of Biomedical Engineering
- The University of Akron
| |
Collapse
|
40
|
Zhang H, Feng G, Guo Y, Zhou D. Robust and specific ratiometric biosensing using a copper-free clicked quantum dot-DNA aptamer sensor. NANOSCALE 2013; 5:10307-15. [PMID: 24056667 PMCID: PMC3814187 DOI: 10.1039/c3nr02897f] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/13/2013] [Indexed: 05/30/2023]
Abstract
We report herein the successful preparation of a compact and functional CdSe-ZnS core-shell quantum dot (QD)-DNA conjugate via highly efficient copper-free "click chemistry" (CFCC) between a dihydro-lipoic acid-polyethylene glycol-azide (DHLA-PEG-N3) capped QD and a cyclooctyne modified DNA. This represents an excellent balance between the requirements of high sensitivity, robustness and specificity for the QD-FRET (Förster resonance energy transfer) based sensor as confirmed by a detailed FRET analysis on the QD-DNA conjugate, yielding a relatively short donor-acceptor distance of ~5.8 nm. We show that this CFCC clicked QD-DNA conjugate is not only able to retain the native fluorescence quantum yield (QY) of the parent DHLA-PEG-N3 capped QD, but also well-suited for robust and specific biosensing; it can directly quantitate, at the pM level, both labelled and unlabelled complementary DNA probes with a good SNP (single-nucleotide polymorphism) discrimination ability in complex media, e.g. 10% human serum via target-binding induced FRET changes between the QD donor and the dye acceptor. Furthermore, this sensor has also been successfully exploited for the detection, at the pM level, of a specific protein target (thrombin) via the encoded anti-thrombin aptamer sequence in the QD-DNA conjugate.
Collapse
Affiliation(s)
- Haiyan Zhang
- School of Chemistry and Astbury Centre for Structural Molecular Biology , University of Leeds , Leeds LS2 9JT , UK . ;
| | - Guoqiang Feng
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education , College of Chemistry , Central China Normal University , 152 Luoyu Road , Wuhan 430079 , P.R. China
| | - Yuan Guo
- School of Chemistry and Astbury Centre for Structural Molecular Biology , University of Leeds , Leeds LS2 9JT , UK . ;
| | - Dejian Zhou
- School of Chemistry and Astbury Centre for Structural Molecular Biology , University of Leeds , Leeds LS2 9JT , UK . ;
| |
Collapse
|
41
|
Thandu M, Rapozzi V, Xodo L, Albericio F, Comuzzi C, Cavalli S. “Clicking” Porphyrins to Magnetic Nanoparticles for Photodynamic Therapy. Chempluschem 2013; 79:90-98. [DOI: 10.1002/cplu.201300276] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Indexed: 11/10/2022]
|
42
|
Development of Magnetic Nanoparticles for Cancer Gene Therapy: A Comprehensive Review. ACTA ACUST UNITED AC 2013. [DOI: 10.1155/2013/646284] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Since they were first proposed as nonviral transfection agents for their gene-carrying capacity, magnetic nanoparticles have been studied thoroughly, both in vitro and in vivo. Great effort has been made to manufacture biocompatible magnetic nanoparticles for use in the theragnosis of cancer and other diseases. Here we survey recent advances in the study of magnetic nanoparticles, as well as the polymers and other coating layers currently available for gene therapy, their synthesis, and bioconjugation processes. In addition, we review several gene therapy models based on magnetic nanoparticles.
Collapse
|
43
|
Avti PK, Maysinger D, Kakkar A. Alkyne-azide "click" chemistry in designing nanocarriers for applications in biology. Molecules 2013; 18:9531-49. [PMID: 23966076 PMCID: PMC6270461 DOI: 10.3390/molecules18089531] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/03/2013] [Accepted: 08/05/2013] [Indexed: 12/11/2022] Open
Abstract
The alkyne-azide cycloaddition, popularly known as the "click" reaction, has been extensively exploited in molecule/macromolecule build-up, and has offered tremendous potential in the design of nanomaterials for applications in a diverse range of disciplines, including biology. Some advantageous characteristics of this coupling include high efficiency, and adaptability to the environment in which the desired covalent linking of the alkyne and azide terminated moieties needs to be carried out. The efficient delivery of active pharmaceutical agents to specific organelles, employing nanocarriers developed through the use of "click" chemistry, constitutes a continuing topical area of research. In this review, we highlight important contributions click chemistry has made in the design of macromolecule-based nanomaterials for therapeutic intervention in mitochondria and lipid droplets.
Collapse
Affiliation(s)
- Pramod K. Avti
- Montreal Heart Institute, Research Center, 5000 Bélanger Est, Montréal, QC H1T 1C8, Canada
- Institute of Biomedical Engineering, École Polytechnique de Montréal, Montreal, QC H3C 3A7, Canada
- Department of Chemistry, McGill University, 801 Sherbrooke St. W. Montréal, QC H3A 0B8 Canada
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke St. W. Montréal, QC H3A 0B8 Canada
| |
Collapse
|
44
|
Arumugam S, Orski SV, Mbua NE, McNitt C, Boons GJ, Locklin J, Popik VV. Photo-click chemistry strategies for spatiotemporal control of metal-free ligation, labeling, and surface derivatization. PURE APPL CHEM 2013. [DOI: 10.1351/pac-con-13-01-08] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Three photo-click ligation strategies described in this account provide
scientists with efficient and selective tools for derivatization of various
molecules, polymers, and surfaces. Fast photochemical reactions that are
utilized in these techniques permit spatiotemporal control of the process. The
absence of activating reagents and catalysts, as well as compatibility with
aqueous media, makes photo-click ligations suitable for biomedical applications.
The first of these approaches relies on the photochemical decarbonylation of
cyclopropenones to produce cyclooctynes. The latter undergo rapid catalyst-free
strain-promoted azide–alkyne cycloaddition (SPAAC) to azide-tagged substrates.
The second method is based on a very fast (>104 M–1
s–1) light-triggered hetero-Diels–Alder reaction and permits
efficient derivatization of substrates bearing vinyl ether moiety. An even
faster reaction between photochemically generated naphthoquinone methides
(oNQMs) and thiols (~2 × 105 M–1
s–1) serves as a basis for a third method. This thiol photo-click
chemistry allows for the selective derivatization of thiol-functionalized
substrates or labeling of free cysteine residues in proteins. The thioether
linkage produced by the reaction of oNQMs and a thiol is stable
under ambient conditions, but can be cleaved by UV irradiation, regenerating
free thiol. This feature permits the removal or replacement of immobilized
compounds, as well as traceless substrate release.
Collapse
Affiliation(s)
| | - Sara V. Orski
- 1Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Ngalle Eric Mbua
- 3Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | | | - Geert-Jan Boons
- 3Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Jason Locklin
- 1Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Vladimir V. Popik
- 1Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
45
|
Peripherally administered nanoparticles target monocytic myeloid cells, secondary lymphoid organs and tumors in mice. PLoS One 2013; 8:e61646. [PMID: 23626707 PMCID: PMC3633981 DOI: 10.1371/journal.pone.0061646] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 03/12/2013] [Indexed: 01/05/2023] Open
Abstract
Nanoparticles have been extensively developed for therapeutic and diagnostic applications. While the focus of nanoparticle trafficking in vivo has traditionally been on drug delivery and organ-level biodistribution and clearance, recent work in cancer biology and infectious disease suggests that targeting different cells within a given organ can substantially affect the quality of the immunological response. Here, we examine the cell-level biodistribution kinetics after administering ultrasmall Pluronic-stabilized poly(propylene sulfide) nanoparticles in the mouse. These nanoparticles depend on lymphatic drainage to reach the lymph nodes and blood, and then enter the spleen rather than the liver, where they interact with monocytes, macrophages and myeloid dendritic cells. They were more readily taken up into lymphatics after intradermal (i.d.) compared to intramuscular administration, leading to ∼50% increased bioavailability in blood. When administered i.d., their distribution favored antigen-presenting cells, with especially strong targeting to myeloid cells. In tumor-bearing mice, the monocytic and the polymorphonuclear myeloid-derived suppressor cell compartments were efficiently and preferentially targeted, rendering this nanoparticulate formulation potentially useful for reversing the highly suppressive activity of these cells in the tumor stroma.
Collapse
|
46
|
|
47
|
Sapsford KE, Algar WR, Berti L, Gemmill KB, Casey BJ, Oh E, Stewart MH, Medintz IL. Functionalizing nanoparticles with biological molecules: developing chemistries that facilitate nanotechnology. Chem Rev 2013; 113:1904-2074. [PMID: 23432378 DOI: 10.1021/cr300143v] [Citation(s) in RCA: 824] [Impact Index Per Article: 74.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Kim E Sapsford
- Division of Biology, Department of Chemistry and Materials Science, Office of Science and Engineering Laboratories, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Gallo J, Long NJ, Aboagye EO. Magnetic nanoparticles as contrast agents in the diagnosis and treatment of cancer. Chem Soc Rev 2013; 42:7816-33. [DOI: 10.1039/c3cs60149h] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
49
|
Pan H, Myerson JW, Hu L, Marsh JN, Hou K, Scott MJ, Allen JS, Hu G, San Roman S, Lanza GM, Schreiber RD, Schlesinger PH, Wickline SA. Programmable nanoparticle functionalization for in vivo targeting. FASEB J 2012; 27:255-64. [PMID: 23047896 DOI: 10.1096/fj.12-218081] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The emerging demand for programmable functionalization of existing base nanocarriers necessitates development of an efficient approach for cargo loading that avoids nanoparticle redesign for each individual application. Herein, we demonstrate in vivo a postformulation strategy for lipidic nanocarrier functionalization with the use of a linker peptide, which rapidly and stably integrates cargos into lipidic membranes of nanocarriers after simple mixing through a self-assembling process. We exemplified this strategy by generating a VCAM-1-targeted perfluorocarbon nanoparticle for in vivo targeting in atherosclerosis (ApoE-deficient) and breast cancer (STAT-1-deficient) models. In the atherosclerotic model, a 4.1-fold augmentation in binding to affected aortas was observed for targeted vs. nontargeted nanoparticles (P<0.0298). Likewise, in the breast cancer model, a 4.9-fold increase in the nanoparticle signal from tumor vasculature was observed for targeted vs. nontargeted nanoparticles (P<0.0216). In each case, the nanoparticle was registered with fluorine ((19)F) magnetic resonance spectroscopy of the nanoparticle perfluorocarbon core, yielding a quantitative estimate of the number of tissue-bound nanoparticles. Because other common nanocarriers with lipid coatings (e.g., liposomes, micelles, etc.) can employ this strategy, this peptide linker postformulation approach is applicable to more than half of the available nanosystems currently in clinical trials or clinical uses.
Collapse
Affiliation(s)
- Hua Pan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63108, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Schieber C, Bestetti A, Lim JP, Ryan AD, Nguyen TL, Eldridge R, White AR, Gleeson PA, Donnelly PS, Williams SJ, Mulvaney P. Conjugation of transferrin to azide-modified CdSe/ZnS core-shell quantum dots using cyclooctyne click chemistry. Angew Chem Int Ed Engl 2012; 51:10523-7. [PMID: 22996637 DOI: 10.1002/anie.201202876] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Revised: 07/05/2012] [Indexed: 12/18/2022]
Abstract
Twinkle twinkle quantum dot: Conjugation of biomolecules to azide-modified quantum dots (QDs) through a bifunctional linker, using strain-promoted azide-alkyne cycloaddition with the QD and a squaramide linkage to the biomolecule (see scheme). Transferrin-conjugated QDs were internalized by transferrin-receptor expressing HeLa cells.
Collapse
Affiliation(s)
- Christine Schieber
- School of Chemistry and Bio21 Institute, University of Melbourne, Parkville, Vic 3010, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|