51
|
Huang L, Leung PKK, Lee LCC, Xu GX, Lam YW, Lo KKW. Photofunctional cyclometallated iridium(III) polypyridine methylsulfone complexes as sulfhydryl-specific reagents for bioconjugation, bioimaging and photocytotoxic applications. Chem Commun (Camb) 2022; 58:10162-10165. [PMID: 35997227 DOI: 10.1039/d2cc02405e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report herein near-infrared (NIR)-emitting cyclometallated iridium(III) complexes bearing a heteroaromatic methylsulfone moiety as sulfhydryl-specific reagents; one of the complexes was conjugated to cysteine and cysteine-containing peptides and proteins for bioimaging and photocytotoxic applications.
Collapse
Affiliation(s)
- Lili Huang
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Peter Kam-Keung Leung
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China. .,State Key Laboratory of Terahertz and Millimetre Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| | - Lawrence Cho-Cheung Lee
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China. .,Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503 - 1511, 15/F, Building 17W, Hong Kong Science Park, New Territories, Hong Kong, P. R. China
| | - Guang-Xi Xu
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Yun-Wah Lam
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China.
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong, P. R. China. .,State Key Laboratory of Terahertz and Millimetre Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| |
Collapse
|
52
|
Judmann B, Braun D, Schirrmacher R, Wängler B, Fricker G, Wängler C. Toward the Development of GE11-Based Radioligands for Imaging of Epidermal Growth Factor Receptor-Positive Tumors. ACS OMEGA 2022; 7:27690-27702. [PMID: 35967067 PMCID: PMC9366781 DOI: 10.1021/acsomega.2c03407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
The epidermal growth factor receptor (EGFR) is closely associated with tumor development and progression and thus an important target structure for imaging and therapy of various tumors. As a result of its important role in malignancies of various origins and the fact that antibody-based compounds targeting the EGFR have significant drawbacks in terms of in vivo pharmacokinetics, several attempts have been made within the last five years to develop peptide-based EGFR-specific radioligands based on the GE11 scaffold. However, none of these approaches have shown convincing results so far, which has been proposed to be attributed to different potential challenges associated with the GE11 lead structure: first, an aggregation of radiolabeled peptides, which might prevent their interaction with their target receptor, or second, a relatively low affinity of monomeric GE11, necessitating its conversion into a multimeric or polymeric form to achieve adequate EGFR-targeting properties. In the present work, we investigated if these aforementioned points are indeed critical and if the EGFR-targeting ability of GE11 can be improved by choosing an appropriate hydrophilic molecular design or a peptide multimer system to obtain a promising radiopeptide for the visualization of EGFR-overexpressing malignancies by positron emission tomography (PET). For this purpose, we developed several monovalent 68Ga-labeled GE11-based agents, a peptide homodimer and a homotetramer to overcome the challenges associated with GE11. The developed ligands were successfully labeled with 68Ga3+ in high radiochemical yields of ≥97% and molar activities of 41-104 GBq/μmol. The resulting radiotracers presented log D(7.4) values between -2.17 ± 0.21 and -3.79 ± 0.04 as well as a good stability in human serum with serum half-lives of 112 to 217 min for the monovalent radiopeptides and 84 and 62 min for the GE11 homodimer and homotetramer, respectively. In the following in vitro studies, none of the 68Ga-labeled radiopeptides demonstrated a considerable EGF receptor-specific uptake in EGFR-positive A431 cells. Moreover, none of the agents was able to displace [125I]I-EGF from the EGFR in competitive displacement assays in the same cell line in concentrations of up to 1 mM, whereas the endogenous receptor ligand hEGF demonstrated a high affinity of 15.2 ± 3.3 nM. These results indicate that it is not the aggregation of the GE11 sequence that seems to be the factor limiting the usefulness of the peptide as basis for radiotracer design but the limited affinity of monovalent and small homomultivalent GE11-based radiotracers to the EGFR. This highlights that the development of small-molecule GE11-based radioligands is not promising.
Collapse
Affiliation(s)
- Benedikt Judmann
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Diana Braun
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Ralf Schirrmacher
- Department
of Oncology, Division of Oncological Imaging, University of Alberta, 11560 University Avenue, T6G 1Z2 Edmonton, AB, Canada
| | - Björn Wängler
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Gert Fricker
- Institute
of Pharmacy and Molecular Biotechnology, University of Heidelberg, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| | - Carmen Wängler
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| |
Collapse
|
53
|
Challenges for the application of EGFR-targeting peptide GE11 in tumor diagnosis and treatment. J Control Release 2022; 349:592-605. [PMID: 35872181 DOI: 10.1016/j.jconrel.2022.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 11/20/2022]
Abstract
Abnormal regulation of cell signaling pathways on cell survival, proliferation and migration contributes to the development of malignant tumors. Among them, epidermal growth factor receptor (EGFR) is one of the most important biomarkers in many types of malignant solid tumors. Its over-expression and mutation status can be served as a biomarker to identify patients who can be benifit from EGFR tyrosine kinase inhibitors and anti-EGFR monocloncal antibody (mAb) therapy. For decades, researches on EGFR targeted ligands were actively carried out to identify potent candidates for cancer therapy. An ideal EGFR ligand can competitively inhibit the binding of endogenous growth factor, such as epidermal growth factor (EGF) and transforming growth factor-α(TGF-α) to EGFR, thus block EGFR signaling pathway and downregulate EGFR expression. Alternatively, conjugation of EGFR ligands on drug delivery systems (DDS) can facilitate targeting delivery of therapeutics or diagnostic agents to EGFR over-expression tumors via EGFR-mediated endocytosis. GE11 peptide is one of the potent EGFR ligand screened from a phage display peptide library. It is a dodecapeptide that can specifically binds to EGFR with high affinity and selectivity. GE11 has been widely used in the diagnosis and targeted delivery of drugs for radiotherapy, genetherapy and chemotherpy against EGFR positive tumors. In this review, the critical factors affecting the in vivo and in vitro targeting performance of GE11 peptide, including ligand-receptor intermolecular force, linker bond properties and physiochemical properties of carrier materials, are detailedly interpreted. This review provides a valuable vision for the rational design and optimization of GE11-based active targeting strategies for cancer treatment, and it will promote the translation studies of GE11 from lab research to clinical application.
Collapse
|
54
|
Furman O, Zaporozhets A, Tobi D, Bazylevich A, Firer MA, Patsenker L, Gellerman G, Lubin BCR. Novel Cyclic Peptides for Targeting EGFR and EGRvIII Mutation for Drug Delivery. Pharmaceutics 2022; 14:1505. [PMID: 35890400 PMCID: PMC9318536 DOI: 10.3390/pharmaceutics14071505] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 02/05/2023] Open
Abstract
The epidermal growth factor-epidermal growth factor receptor (EGF-EGFR) pathway has become the main focus of selective chemotherapeutic intervention. As a result, two classes of EGFR inhibitors have been clinically approved, namely monoclonal antibodies and small molecule kinase inhibitors. Despite an initial good response rate to these drugs, most patients develop drug resistance. Therefore, new treatment approaches are needed. In this work, we aimed to find a new EGFR-specific, short cyclic peptide, which could be used for targeted drug delivery. Phage display peptide technology and biopanning were applied to three EGFR expressing cells, including cells expressing the EGFRvIII mutation. DNA from the internalized phage was extracted and the peptide inserts were sequenced using next-generation sequencing (NGS). Eleven peptides were selected for further investigation using binding, internalization, and competition assays, and the results were confirmed by confocal microscopy and peptide docking. Among these eleven peptides, seven showed specific and selective binding and internalization into EGFR positive (EGFR+ve) cells, with two of them-P6 and P9-also demonstrating high specificity for non-small cell lung cancer (NSCLC) and glioblastoma cells, respectively. These peptides were chemically conjugated to camptothecin (CPT). The conjugates were more cytotoxic to EGFR+ve cells than free CPT. Our results describe a novel cyclic peptide, which can be used for targeted drug delivery to cells overexpressing the EGFR and EGFRvIII mutation.
Collapse
Affiliation(s)
- Olga Furman
- Department of Chemical Engineering, Biotechnology and Materials, Ariel University, Ariel 40700, Israel; (O.F.); (M.A.F.)
- Agriculture and Oenology Department, Eastern Regional R&D Center, Ariel 40700, Israel
| | - Alisa Zaporozhets
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.Z.); (A.B.); (L.P.); (G.G.)
| | - Dror Tobi
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
- Department of Molecular Biology, Ariel University, Ariel 40700, Israel
| | - Andrii Bazylevich
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.Z.); (A.B.); (L.P.); (G.G.)
| | - Michael A. Firer
- Department of Chemical Engineering, Biotechnology and Materials, Ariel University, Ariel 40700, Israel; (O.F.); (M.A.F.)
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
- Ariel Center for Applied Cancer Research, Ariel 40700, Israel
| | - Leonid Patsenker
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.Z.); (A.B.); (L.P.); (G.G.)
| | - Gary Gellerman
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.Z.); (A.B.); (L.P.); (G.G.)
- Ariel Center for Applied Cancer Research, Ariel 40700, Israel
| | - Bat Chen R. Lubin
- Department of Chemical Engineering, Biotechnology and Materials, Ariel University, Ariel 40700, Israel; (O.F.); (M.A.F.)
- Agriculture and Oenology Department, Eastern Regional R&D Center, Ariel 40700, Israel
| |
Collapse
|
55
|
Liu C, Zhao Z, Gao R, Zhang X, Sun Y, Wu J, Liu J, Chen C. Matrix Metalloproteinase-2-Responsive Surface-Changeable Liposomes Decorated by Multifunctional Peptides to Overcome the Drug Resistance of Triple-Negative Breast Cancer through Enhanced Targeting and Penetrability. ACS Biomater Sci Eng 2022; 8:2979-2994. [PMID: 35666956 DOI: 10.1021/acsbiomaterials.2c00295] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although nanomedicine has demonstrated great potential for combating drug resistance, its suboptimal recognition of malignant cells and limited transport across multiple biological obstacles seriously impede the efficacious accumulation of drugs in tumor lesions, which strikingly limits its application in the clinical therapy of drug-resistant triple-negative breast cancer (TNBC). Hence, a surface-variable drug delivery vehicle based on the modification of liposomes with a multifunctional peptide (named EMC) was fabricated in this work and used for encapsulating doxorubicin and the p-glycoprotein inhibitor tariquidar. This EMC peptide contains an EGFR-targeting bullet that was screened from a "one-bead one-compound" combinatorial library, an MMP-2-cleaved substrate, and a cell-penetrating segment. The EGFR-targeting sequence has been validated to possess excellent specificity and affinity for EGFR at both the cellular and molecular levels and could be unloaded from the EMC peptide by MMP-2 in the tumor microenvironment. This doxorubicin/tariquidar-coloaded and peptide-functionalized liposome (DT-pLip) exhibited superior efficacy in tumor growth inhibition to drug-resistant TNBC both in vitro and in vivo through EGFR targeting, osmotic enhancement in response to MMP-2, controllable release, and inhibited efflux. Consequently, our systematic studies indicated the potential of this liposome-based nanoplatform in the therapy of drug-resistant TNBC through targeting effects and tumor microenvironment-triggered penetration enhancement.
Collapse
Affiliation(s)
- Changliang Liu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, National clinical research center for geriatrics, Translational Neuroscience center, Department of Anesthesiology, The Research Units of West China, Chinese Academy of Medical Science, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Zijian Zhao
- BOE Technology Group Co., Ltd., Beijing 100176, China
| | - Rui Gao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, National clinical research center for geriatrics, Translational Neuroscience center, Department of Anesthesiology, The Research Units of West China, Chinese Academy of Medical Science, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Xueying Zhang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, National clinical research center for geriatrics, Translational Neuroscience center, Department of Anesthesiology, The Research Units of West China, Chinese Academy of Medical Science, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yalan Sun
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, National clinical research center for geriatrics, Translational Neuroscience center, Department of Anesthesiology, The Research Units of West China, Chinese Academy of Medical Science, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jiahui Wu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, National clinical research center for geriatrics, Translational Neuroscience center, Department of Anesthesiology, The Research Units of West China, Chinese Academy of Medical Science, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jin Liu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, National clinical research center for geriatrics, Translational Neuroscience center, Department of Anesthesiology, The Research Units of West China, Chinese Academy of Medical Science, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Chan Chen
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, National clinical research center for geriatrics, Translational Neuroscience center, Department of Anesthesiology, The Research Units of West China, Chinese Academy of Medical Science, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
56
|
Del Genio V, Bellavita R, Falanga A, Hervé-Aubert K, Chourpa I, Galdiero S. Peptides to Overcome the Limitations of Current Anticancer and Antimicrobial Nanotherapies. Pharmaceutics 2022; 14:1235. [PMID: 35745807 PMCID: PMC9230615 DOI: 10.3390/pharmaceutics14061235] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/25/2022] [Accepted: 06/09/2022] [Indexed: 12/13/2022] Open
Abstract
Biomedical research devotes a huge effort to the development of efficient non-viral nanovectors (NV) to improve the effectiveness of standard therapies. NVs should be stable, sustainable and biocompatible and enable controlled and targeted delivery of drugs. With the aim to foster the advancements of such devices, this review reports some recent results applicable to treat two types of pathologies, cancer and microbial infections, aiming to provide guidance in the overall design of personalized nanomedicines and highlight the key role played by peptides in this field. Additionally, future challenges and potential perspectives are illustrated, in the hope of accelerating the translational advances of nanomedicine.
Collapse
Affiliation(s)
- Valentina Del Genio
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80138 Naples, Italy; (V.D.G.); (R.B.)
- EA 6295 Nanomédicaments et Nanosondes, University of Tours, UFR Pharmacie, 31 Avenue Monge, 37200 Tours, France;
| | - Rosa Bellavita
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80138 Naples, Italy; (V.D.G.); (R.B.)
| | - Annarita Falanga
- Department of Agricultural Science, University of Naples “Federico II”, Via Università 100, 80055 Naples, Italy;
| | - Katel Hervé-Aubert
- EA 6295 Nanomédicaments et Nanosondes, University of Tours, UFR Pharmacie, 31 Avenue Monge, 37200 Tours, France;
| | - Igor Chourpa
- EA 6295 Nanomédicaments et Nanosondes, University of Tours, UFR Pharmacie, 31 Avenue Monge, 37200 Tours, France;
| | - Stefania Galdiero
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80138 Naples, Italy; (V.D.G.); (R.B.)
| |
Collapse
|
57
|
Tong S, Darwish S, Ariani HHN, Lozada KA, Salehi D, Cinelli MA, Silverman RB, Kaur K, Yang S. A Small Peptide Increases Drug Delivery in Human Melanoma Cells. Pharmaceutics 2022; 14:1036. [PMID: 35631623 PMCID: PMC9145755 DOI: 10.3390/pharmaceutics14051036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 02/04/2023] Open
Abstract
Melanoma is the most fatal type of skin cancer and is notoriously resistant to chemotherapies. The response of melanoma to current treatments is difficult to predict. To combat these challenges, in this study, we utilize a small peptide to increase drug delivery to melanoma cells. A peptide library array was designed and screened using a peptide array-whole cell binding assay, which identified KK-11 as a novel human melanoma-targeting peptide. The peptide and its D-amino acid substituted analogue (VPWxEPAYQrFL or D-aa KK-11) were synthesized via a solid-phase strategy. Further studies using FITC-labeled KK-11 demonstrated dose-dependent uptake in human melanoma cells. D-aa KK-11 significantly increased the stability of the peptide, with 45.3% remaining detectable after 24 h with human serum incubation. Co-treatment of KK-11 with doxorubicin was found to significantly enhance the cytotoxicity of doxorubicin compared to doxorubicin alone, or sequential KK-11 and doxorubicin treatment. In vivo and ex vivo imaging revealed that D-aa KK-11 distributed to xenografted A375 melanoma tumors as early as 5 min and persisted up to 24 h post tail vein injection. When co-administered, D-aa KK-11 significantly enhanced the anti-tumor activity of a novel nNOS inhibitor (MAC-3-190) in an A375 human melanoma xenograft mouse model compared to MAC-3-190 treatment alone. No apparent systemic toxicities were observed. Taken together, these results suggest that KK-11 may be a promising human melanoma-targeted delivery vector for anti-melanoma cargo.
Collapse
Affiliation(s)
- Shirley Tong
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.T.); (K.A.L.)
| | - Shaban Darwish
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.D.); (H.H.N.A.); (D.S.)
| | - Hanieh Hossein Nejad Ariani
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.D.); (H.H.N.A.); (D.S.)
| | - Kate Alison Lozada
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.T.); (K.A.L.)
| | - David Salehi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.D.); (H.H.N.A.); (D.S.)
| | - Maris A. Cinelli
- Center for Developmental Therapeutics, Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; (M.A.C.); (R.B.S.)
| | - Richard B. Silverman
- Center for Developmental Therapeutics, Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; (M.A.C.); (R.B.S.)
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kamaljit Kaur
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.D.); (H.H.N.A.); (D.S.)
| | - Sun Yang
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.T.); (K.A.L.)
| |
Collapse
|
58
|
Decker S, Taschauer A, Geppl E, Pirhofer V, Schauer M, Pöschl S, Kopp F, Richter L, Ecker GF, Sami H, Ogris M. Structure-based peptide ligand design for improved epidermal growth factor receptor targeted gene delivery. Eur J Pharm Biopharm 2022; 176:211-221. [DOI: 10.1016/j.ejpb.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/25/2022] [Accepted: 05/04/2022] [Indexed: 11/04/2022]
|
59
|
Vetter VC, Wagner E. Targeting nucleic acid-based therapeutics to tumors: Challenges and strategies for polyplexes. J Control Release 2022; 346:110-135. [PMID: 35436520 DOI: 10.1016/j.jconrel.2022.04.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/07/2022] [Accepted: 04/10/2022] [Indexed: 12/18/2022]
Abstract
The current medical reality of cancer gene therapy is reflected by more than ten approved products on the global market, including oncolytic and other viral vectors and CAR T-cells as ex vivo gene-modified cell therapeutics. The development of synthetic antitumoral nucleic acid therapeutics has been proceeding at a lower but steady pace, fueled by a plethora of alternative nucleic acid platforms (from various antisense oligonucleotides, siRNA, microRNA, lncRNA, sgRNA, to larger mRNA and DNA) and several classes of physical and chemical delivery technologies. This review summarizes the challenges and strategies for tumor-targeted nucleic acid delivery. Focusing primarily on polyplexes (polycation complexes) as nanocarriers, delivery options across multiple barriers into tumor cells are illustrated.
Collapse
Affiliation(s)
- Victoria C Vetter
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig-Maximilians-Universität, Munich 81377, Germany; Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Munich 81377, Germany.
| |
Collapse
|
60
|
de Paiva IM, Vakili MR, Soleimani AH, Tabatabaei Dakhili SA, Munira S, Paladino M, Martin G, Jirik FR, Hall DG, Weinfeld M, Lavasanifar A. Biodistribution and Activity of EGFR Targeted Polymeric Micelles Delivering a New Inhibitor of DNA Repair to Orthotopic Colorectal Cancer Xenografts with Metastasis. Mol Pharm 2022; 19:1825-1838. [PMID: 35271294 PMCID: PMC9175178 DOI: 10.1021/acs.molpharmaceut.1c00918] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The disruption of polynucleotide kinase/phosphatase (PNKP) in colorectal cancer (CRC) cells deficient in phosphatase and tensin homolog (PTEN) is expected to lead to the loss of cell viability by a process known as synthetic lethality. In previous studies, we have reported on the encapsulation of a novel inhibitor of PNKP, namely, A83B4C63, in polymeric micelles and its activity in slowing the growth of PTEN-deficient CRC cells as well as subcutaneous xenografts. In this study, to enhance drug delivery and specificity to CRC tumors, the surface of polymeric micelles carrying A83B4C63 was modified with GE11, a peptide targeting epidermal growth factor receptor (EGFR) overexpressed in about 70% of CRC tumors. Using molecular dynamics (MD) simulations, we assessed the binding site and affinity of GE11 for EGFR. The GE11-modified micelles, tagged with a near-infrared fluorophore, showed enhanced internalization by EGFR-overexpressing CRC cells in vitro and a trend toward increased primary tumor homing in an orthotopic CRC xenograft in vivo. In line with these observations, the GE11 modification of polymeric micelles was shown to positively contribute to the improved therapeutic activity of encapsulated A83B4C63 against HCT116-PTEN-/- cells in vitro and that of orthotopic CRC xenograft in vivo. In conclusion, our results provided proof of principle evidence for the potential benefit of EGFR targeted polymeric micellar formulations of A83B4C63 as monotherapeutics for aggressive and metastatic CRC tumors but at the same time highlighted the need for the development of EGFR ligands with improved physiological stability and EGFR binding.
Collapse
Affiliation(s)
- Igor Moura de Paiva
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2EZ, Canada
| | - Mohammad Reza Vakili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2EZ, Canada
| | - Amir Hasan Soleimani
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2EZ, Canada
| | | | - Sirazum Munira
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2EZ, Canada
| | - Marco Paladino
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | | | | | - Dennis G Hall
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Michael Weinfeld
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2EZ, Canada.,Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 2H5, Canada
| |
Collapse
|
61
|
Xu X, Jin C, Zhang K, Cao Y, Liu J, Zhang Y, Ran H, Jin Y. Activatable “Matryoshka” nanosystem delivery NgBR siRNA and control drug release for stepwise therapy and evaluate drug resistance cancer. Mater Today Bio 2022; 14:100245. [PMID: 35345559 PMCID: PMC8956824 DOI: 10.1016/j.mtbio.2022.100245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/02/2022] [Accepted: 03/16/2022] [Indexed: 12/03/2022] Open
Abstract
Drug resistance is always a challenge in conquering breast cancer clinically. Recognition of drug resistance and enhancing the sensitivity of the tumor to chemotherapy is urgent. Herein, a dual-responsive multi-function “Matryoshka" nanosystem is designed, it activates in the tumor microenvironment, decomposes layer by layer, and release gene and drug in sequence. The cell is re-educated by NgBR siRNA first to regain the chemosensitivity through regulating the Akt pathway and inhibit ERα activation, then the drugs loaded in the core are controlled released to killing cells. Carbonized polymer dots are loaded into the nanosystem as an efficient bioimaging probe, due to the GE11 modification, the nanosystem can be a seeker to recognize and evaluate drug-resistance tumors by photoacoustic imaging. In the tumor-bearing mouse, the novel nanosystem firstly enhances the sensitivity to chemotherapy by knockdown NgBR, inducing a much higher reduction in NgBR up to 52.09%, then effectively inhibiting tumor growth by chemotherapy, tumor growth in nude mouse was inhibited by 70.22%. The nanosystem also can inhibit metastasis, prolong survival time, and evaluate tumor drug resistance by real-time imaging. Overall, based on regulating the key molecules of drug resistance, we created visualization nanotechnology and formatted new comprehensive plans with high bio-safety for tumor diagnosis and treatment, providing a personalized strategy to overcome drug resistance clinically. Knockdown NgBR regulate the Akt pathway and inhibit ERα activate, enhance the sensitivity of chemotherapy. Knockdown of NgBR inhibits metastasis and prolongs survival. Nanosystem can evaluate drug resistance and kill tumors at the same time.
Collapse
|
62
|
Zhao J, Li S, Pang X, Shan Y. Evaluating the therapeutic efficacy of nano-drugs targeting epidermal growth factor receptor. Chem Commun (Camb) 2022; 58:2726-2729. [PMID: 35113095 DOI: 10.1039/d1cc06754k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Epidermal growth factor receptor (EGFR) targeted nano-drugs facilitate effective diagnosis and treatment of cancer. Herein, the therapeutic efficacy of nano-drugs targeting EGFR was evaluated from the perspective of cell entry efficiency and induced cell mechanical properties using force tracing and nano-indentation techniques at the single particle/cell level in real time.
Collapse
Affiliation(s)
- Jing Zhao
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China.
| | - Siying Li
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China.
| | - Xuelei Pang
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China.
| | - Yuping Shan
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China.
| |
Collapse
|
63
|
Kanada M, Linenfelser L, Cox E, Gilad AA. A Dual-Reporter Platform for Screening Tumor-Targeted Extracellular Vesicles. Pharmaceutics 2022; 14:475. [PMID: 35335849 PMCID: PMC8953635 DOI: 10.3390/pharmaceutics14030475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/10/2022] [Accepted: 02/18/2022] [Indexed: 12/10/2022] Open
Abstract
Extracellular vesicle (EV)-mediated transfer of biomolecules plays an essential role in intercellular communication and may improve targeted drug delivery. In the past decade, various approaches to EV surface modification for targeting specific cells or tissues have been proposed, including genetic engineering of parental cells or postproduction EV engineering. However, due to technical limitations, targeting moieties of engineered EVs have not been thoroughly characterized. Here, we report the bioluminescence resonance energy transfer (BRET) EV reporter, PalmReNL-based dual-reporter platform for characterizing the cellular uptake of tumor-homing peptide (THP)-engineered EVs, targeting PDL1, uPAR, or EGFR proteins expressed in MDA-MB-231 breast cancer cells, simultaneously by bioluminescence measurement and fluorescence microscopy. Bioluminescence analysis of cellular EV uptake revealed the highest binding efficiency of uPAR-targeted EVs, whereas PDL1-targeted EVs showed slower cellular uptake. EVs engineered with two known EGFR-binding peptides via lipid nanoprobes did not increase cellular uptake, indicating that designs of EGFR-binding peptide conjugation to the EV surface are critical for functional EV engineering. Fluorescence analysis of cellular EV uptake allowed us to track individual PalmReNL-EVs bearing THPs in recipient cells. These results demonstrate that the PalmReNL-based EV assay platform can be a foundation for high-throughput screening of tumor-targeted EVs.
Collapse
Affiliation(s)
- Masamitsu Kanada
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA; (L.L.); (E.C.)
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Lauren Linenfelser
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA; (L.L.); (E.C.)
- Department of Chemical Engineering & Materials Science, Michigan State University, East Lansing, MI 48824, USA
| | - Elyssa Cox
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA; (L.L.); (E.C.)
| | - Assaf A. Gilad
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA; (L.L.); (E.C.)
- Department of Chemical Engineering & Materials Science, Michigan State University, East Lansing, MI 48824, USA
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
64
|
Lieser RM, Hartzell EJ, Yur D, Sullivan MO, Chen W. EGFR Ligand Clustering on E2 Bionanoparticles for Targeted Delivery of Chemotherapeutics to Breast Cancer Cells. Bioconjug Chem 2022; 33:452-462. [PMID: 35167278 DOI: 10.1021/acs.bioconjchem.1c00579] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Naturally occurring protein nanocages are promising drug carriers because of their uniform size and biocompatibility. Engineering efforts have enhanced the delivery properties of nanocages, but cell specificity and high drug loading remain major challenges. Herein, we fused the SpyTag peptide to the surface of engineered E2 nanocages to enable tunable nanocage decoration and effective E2 cell targeting using a variety of SpyCatcher (SC) fusion proteins. Additionally, the core of the E2 nanocage incorporated four phenylalanine mutations previously shown to allow hydrophobic loading of doxorubicin and pH-responsive release in acidic environments. We functionalized the surface of the nanocage with a highly cell-specific epidermal growth factor receptor (EGFR)-targeting protein conjugate, 4GE11-mCherry-SC, developed previously in our laboratories by employing unnatural amino acid (UAA) protein engineering chemistries. Herein, we demonstrated the benefits of this engineered protein nanocage construct for efficient drug loading, with a straightforward method for removal of the unloaded drug through elastin-like polypeptide-mediated inverse transition cycling. Additionally, we demonstrated approximately 3-fold higher doxorubicin internalization in inflammatory breast cancer cells compared to healthy breast epithelial cells, leading to targeted cell death at concentrations below the IC50 of free doxorubicin. Collectively, these results demonstrated the versatility of our UAA-based EGFR-targeting protein construct to deliver a variety of cargoes efficiently, including engineered E2 nanocages capable of site-specific functionalization and doxorubicin loading.
Collapse
Affiliation(s)
- Rachel M Lieser
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States
| | - Emily J Hartzell
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States
| | - Daniel Yur
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States
| |
Collapse
|
65
|
Jeong W, Bu J, Jafari R, Rehak P, Kubiatowicz LJ, Drelich AJ, Owen RH, Nair A, Rawding PA, Poellmann MJ, Hopkins CM, Král P, Hong S. Hierarchically Multivalent Peptide-Nanoparticle Architectures: A Systematic Approach to Engineer Surface Adhesion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103098. [PMID: 34894089 PMCID: PMC8811846 DOI: 10.1002/advs.202103098] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/04/2021] [Indexed: 05/20/2023]
Abstract
The multivalent binding effect has been the subject of extensive studies to modulate adhesion behaviors of various biological and engineered systems. However, precise control over the strong avidity-based binding remains a significant challenge. Here, a set of engineering strategies are developed and tested to systematically enhance the multivalent binding of peptides in a stepwise manner. Poly(amidoamine) (PAMAM) dendrimers are employed to increase local peptide densities on a substrate, resulting in hierarchically multivalent architectures (HMAs) that display multivalent dendrimer-peptide conjugates (DPCs) with various configurations. To control binding behaviors, effects of the three major components of the HMAs are investigated: i) poly(ethylene glycol) (PEG) linkers as spacers between conjugated peptides; ii) multiple peptides on the DPCs; and iii) various surface arrangements of HMAs (i.e., a mixture of DPCs each containing different peptides vs DPCs cofunctionalized with multiple peptides). The optimized HMA configuration enables significantly enhanced target cell binding with high selectivity compared to the control surfaces directly conjugated with peptides. The engineering approaches presented herein can be applied individually or in combination, providing guidelines for the effective utilization of biomolecular multivalent interactions using DPC-based HMAs.
Collapse
Affiliation(s)
- Woo‐jin Jeong
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
- Department of Biological Sciences and BioengineeringInha University100 Inha‐ro, Michuhol‐guIncheon22212Republic of Korea
| | - Jiyoon Bu
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Roya Jafari
- Department of ChemistryUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
| | - Pavel Rehak
- Department of ChemistryUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
| | - Luke J. Kubiatowicz
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Adam J. Drelich
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Randall H. Owen
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Ashita Nair
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Piper A. Rawding
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Michael J. Poellmann
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Caroline M. Hopkins
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Petr Král
- Department of ChemistryUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
- Departments of Physics, Pharmaceutical Sciences and Chemical EngineeringUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
- Department of Biomedical EngineeringThe University of Wisconsin‐Madison1550 Engineering Dr.MadisonWI53705USA
- Yonsei Frontier LabDepartment of PharmacyYonsei University50 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| |
Collapse
|
66
|
Zhang X, Chai Z, Lee Dobbins A, Itano MS, Askew C, Miao Z, Niu H, Samulski RJ, Li C. Customized blood-brain barrier shuttle peptide to increase AAV9 vector crossing the BBB and augment transduction in the brain. Biomaterials 2022; 281:121340. [PMID: 34998171 PMCID: PMC8810684 DOI: 10.1016/j.biomaterials.2021.121340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/24/2021] [Accepted: 12/25/2021] [Indexed: 02/03/2023]
Abstract
Recombinant adeno-associated virus (rAAV) vectors have been widely used as favored delivery vehicles for the treatment of inherited diseases in clinical trials, including neurological diseases. However, the noninvasive systemic delivery of rAAV to the central nervous system is severely hampered by the blood-brain barrier (BBB). Several approaches have been exploited to enhance AAV vector brain transduction after systemic administration, including genetic modification of AAV capsids and physical methods. However, these approaches are not always predictive of desirable outcomes in humans and induce complications. It is imperative to explore novel strategies to increase the ability of AAV9 to cross the BBB for enhanced brain transduction. Herein, we have conducted a combinatorial in vivo/in vitro phage display library screening in mouse brains and purified AAV9 virions to identify a customized BBB shuttle peptide, designated as PB5-3. The PB5-3 peptide specifically bound to AAV9 virions and enhanced widespread transduction of AAV9 in mouse brains, especially in neuronal cells, after systemic administration. Further study demonstrated that systemic administration of AAV9 vectors encoding IDUA complexed with PB5-3 increased the phenotypic correction in the brains of MPS I mice. Mechanistic studies revealed that the PB5-3 peptide effectively increased AAV9 trafficking and transcytosis efficiency in the human BBB model hCMEC/D3 cell line but did not interfere with AAV9 binding to the receptor terminal N-linked galactosylated glycans. Additionally, the PB5-3 peptide slowed the clearance of AAV9 from blood without hepatic toxicity. This study highlights, for the first time, the potential of this combinatorial approach for the isolation of peptides that interact with specific AAV vectors for enhanced and targeted AAV transduction. This promising approach will open new combined therapeutic avenues and shed light on the potential applications of peptides for the treatment of human diseases in future clinical trials with AAV vector-mediated gene delivery.
Collapse
Affiliation(s)
- Xintao Zhang
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zheng Chai
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amanda Lee Dobbins
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michelle S Itano
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charles Askew
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zhe Miao
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hongqian Niu
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pharmacology, USA
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
67
|
Lieser RM, Li Q, Chen W, Sullivan MO. Incorporation of Endosomolytic Peptides with Varying Disruption Mechanisms into EGFR-Targeted Protein Conjugates: The Effect on Intracellular Protein Delivery and EGFR Specificity in Breast Cancer Cells. Mol Pharm 2022; 19:661-673. [PMID: 35040326 DOI: 10.1021/acs.molpharmaceut.1c00788] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intracellular delivery of protein therapeutics remains a significant challenge limiting the majority of clinically available protein drugs to extracellular targets. Strategies to deliver proteins to subcellular compartments have traditionally relied on cell-penetrating peptides, which can drive enhanced internalization but exhibit unreliable activity and are rarely able to target specific cells, leading to off-target effects. Moreover, few design rules exist regarding the relative efficacy of various endosomal escape strategies in proteins. Accordingly, we developed a simple fusion modification approach to incorporate endosomolytic peptides onto epidermal growth factor receptor (EGFR)-targeted protein conjugates and performed a systematic comparison of the endosomal escape efficacy, mechanism of action, and capacity to maintain EGFR-targeting specificity of conjugates modified with four different endosomolytic sequences of varying modes of action (Aurein 1.2, GALA, HA2, and L17E). Use of the recently developed Gal8-YFP assay indicated that the fusion of each endosomolytic peptide led to enhanced endosomal disruption. Additionally, the incorporation of each endosomolytic peptide increased the half-life of the internalized protein and lowered lysosomal colocalization, further supporting the membrane-disruptive capacity. Despite this, only EGFR-targeted conjugates modified with Aurein 1.2 or GALA maintained EGFR specificity. These results thus demonstrated that the choice of endosomal escape moiety can substantially affect targeting capability, cytotoxicity, and bioactivity and provided important new insights into endosomolytic peptide selection for the design of targeted protein delivery systems.
Collapse
Affiliation(s)
- Rachel M Lieser
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States
| | - Qirun Li
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States
| |
Collapse
|
68
|
Bashant MM, Mitchell SM, Hart LR, Lebedenko CG, Banerjee IA. In silico studies of interactions of peptide-conjugated cholesterol metabolites and betulinic acid with EGFR, LDR, and N-terminal fragment of CCKA receptors. J Mol Model 2021; 28:16. [PMID: 34961887 DOI: 10.1007/s00894-021-05007-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/14/2021] [Indexed: 11/25/2022]
Abstract
In this work, we designed three new ligands by conjugating cholesterol metabolites 3-hydroxy-5-cholestenoic acid (3-HC) and 3-oxo-4-cholestenoic acid (3-OC) and the natural tri-terpenoid betulinic acid with the tumor-targeting peptide YHWYGYTPQNVI. Molecular interactions with the unconjugated peptide and the conjugates were examined with three receptors that are commonly overexpressed in pancreatic adenocarcinoma cells using ligand docking and molecular dynamics. This study demonstrated the utility of the designed conjugates as a valuable scaffold for potentially targeting EGFR and LDLR receptors. Our results indicate that the conjugates showed strong binding affinities and formation of stable complexes with EGFR, while the unconjugated peptide, BT-peptide conjugate, an 3-HC-peptide conjugate showed the formation of fairly stable complexes with LDLR receptor. For EGFR, two receptor kinase domains were explored. Interactions with the N-terminal domain of CCKA-R were relatively weaker. For LDLR, binding occurred in the beta-propeller region. For the N-terminal fragment of CCKA-R, the conjugates induced significant conformational changes in the receptor. The molecular dynamic simulations for 100 ns demonstrate that BT-peptide conjugates and the unconjugated peptide had the highest binding and formed the most stable complexes with EGFR. RMSD and trajectory analyses indicate that these molecules transit to a dynamically stable configuration in most cases within 60 ns. NMA analysis indicated that amongst the conjugates that showed relatively higher interactions with the respective receptors, the highest potential for deformability was seen for the N-terminal-47 amino acid region of the CCKA-R receptor with and the lowest for the LDLR-receptor. Thus, the newly designed compounds may be evaluated in the future toward developing drug delivery materials for targeting tumor cells overexpressing LDLR or EGFR.
Collapse
Affiliation(s)
- Madeline M Bashant
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Saige M Mitchell
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Lucy R Hart
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Charlotta G Lebedenko
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Ipsita A Banerjee
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA.
| |
Collapse
|
69
|
Spellerberg R, Benli-Hoppe T, Kitzberger C, Berger S, Schmohl KA, Schwenk N, Yen HY, Zach C, Schilling F, Weber WA, Kälin RE, Glass R, Nelson PJ, Wagner E, Spitzweg C. Selective sodium iodide symporter ( NIS) genetherapy of glioblastoma mediatedby EGFR-targeted lipopolyplexes. Mol Ther Oncolytics 2021; 23:432-446. [PMID: 34853814 PMCID: PMC8604759 DOI: 10.1016/j.omto.2021.10.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/11/2021] [Accepted: 10/26/2021] [Indexed: 12/18/2022] Open
Abstract
Lipo-oligomers, post-functionalized with ligands to enhance targeting, represent promising new vehicles for the tumor-specific delivery of therapeutic genes such as the sodium iodide symporter (NIS). Due to its iodide trapping activity, NIS is a powerful theranostic tool for diagnostic imaging and the application of therapeutic radionuclides. 124I PET imaging allows non-invasive monitoring of the in vivo biodistribution of functional NIS expression, and application of 131I enables cytoreduction. In our experimental design, we used epidermal growth factor receptor (EGFR)-targeted polyplexes (GE11) initially characterized in vitro using 125I uptake assays. Mice bearing an orthotopic glioblastoma were treated subsequently with mono-dibenzocyclooctyne (DBCO)-PEG24-GE11/NIS or bisDBCO-PEG24-GE11/NIS, and 24-48 h later, 124I uptake was assessed by positron emission tomography (PET) imaging. The best-performing polyplex in the imaging studies was then selected for 131I therapy studies. The in vitro studies showed EGFR-dependent and NIS-specific transfection efficiency of the polyplexes. The injection of monoDBCO-PEG24-GE11/NIS polyplexes 48 h before 124I application was characterized to be the optimal regime in the imaging studies and was therefore used for an 131I therapy study, showing a significant decrease in tumor growth and a significant extension of survival in the therapy group. These studies demonstrate the potential of EGFR-targeted polyplex-mediated NIS gene therapy as a new strategy for the therapy of glioblastoma.
Collapse
Affiliation(s)
- Rebekka Spellerberg
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Teoman Benli-Hoppe
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, 81377 Munich, Germany
| | - Carolin Kitzberger
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, 81377 Munich, Germany
| | - Kathrin A Schmohl
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Nathalie Schwenk
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Hsi-Yu Yen
- Institute of Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Christian Zach
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Franz Schilling
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
| | - Roland E Kälin
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany.,Walter Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Rainer Glass
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany.,Walter Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, 81377 Munich, Germany.,German Cancer Consortium (DKTK), partner site 80336 Munich and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Peter J Nelson
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, 81377 Munich, Germany
| | - Christine Spitzweg
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany.,Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
70
|
He J, Ren W, Wang W, Han W, Jiang L, Zhang D, Guo M. Exosomal targeting and its potential clinical application. Drug Deliv Transl Res 2021; 12:2385-2402. [PMID: 34973131 PMCID: PMC9458566 DOI: 10.1007/s13346-021-01087-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 12/12/2022]
Abstract
Exosomes are extracellular vesicles secreted by a variety of living cells, which have a certain degree of natural targeting as nano-carriers. Almost all exosomes released by cells will eventually enter the blood circulation or be absorbed by other cells. Under the action of content sorting mechanism, some specific surface molecules can be expressed on the surface of exosomes, such as tetraspanins protein and integrin. To some extent, these specific surface molecules can fuse with specific cells, so that exosomes show specific cell natural targeting. In recent years, exosomes have become a drug delivery system with low immunogenicity, high biocompatibility and high efficacy. Nucleic acids, polypeptides, lipids, or small molecule drugs with therapeutic function are organically loaded into exosomes, and then transported to specific types of cells or tissues in vivo, especially tumor tissues, to achieve targeting drug delivery. The natural targeting of exosome has been found and recognized in some studies, but there are still many challenges in effective clinical treatments. The use of the natural targeting of exosomes alone is incapable of accurately transporting the goods loaded to specific sites. Besides, the natural targeting of exosomes is still an open question in disease targeting and efficient gene/chemotherapy combined therapy. Engineering transformation and modification on exosomes can optimize its natural targeting and deliver the goods to a specific location, providing wide use in clinical treatment. This review summarizes the research progress of exosomal natural targeting and transformation strategy of obtained targeting after transformation. The mechanism of natural targeting and obtained targeting after transformation are also reviewed. The potential value of exosomal targeting in clinical application is also discussed.
Collapse
Affiliation(s)
- Jiao He
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China
| | - Weihong Ren
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, People's Republic of China.
| | - Wei Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, People's Republic of China
| | - Wenyan Han
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, People's Republic of China
| | - Lu Jiang
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, People's Republic of China
| | - Dai Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, People's Republic of China
| | - Mengqi Guo
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
71
|
Pre-Clinical and Clinical Applications of Small Interfering RNAs (siRNA) and Co-Delivery Systems for Pancreatic Cancer Therapy. Cells 2021; 10:cells10123348. [PMID: 34943856 PMCID: PMC8699513 DOI: 10.3390/cells10123348] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer (PC) is one of the leading causes of death and is the fourth most malignant tumor in men. The epigenetic and genetic alterations appear to be responsible for development of PC. Small interfering RNA (siRNA) is a powerful genetic tool that can bind to its target and reduce expression level of a specific gene. The various critical genes involved in PC progression can be effectively targeted using diverse siRNAs. Moreover, siRNAs can enhance efficacy of chemotherapy and radiotherapy in inhibiting PC progression. However, siRNAs suffer from different off target effects and their degradation by enzymes in serum can diminish their potential in gene silencing. Loading siRNAs on nanoparticles can effectively protect them against degradation and can inhibit off target actions by facilitating targeted delivery. This can lead to enhanced efficacy of siRNAs in PC therapy. Moreover, different kinds of nanoparticles such as polymeric nanoparticles, lipid nanoparticles and metal nanostructures have been applied for optimal delivery of siRNAs that are discussed in this article. This review also reveals that how naked siRNAs and their delivery systems can be exploited in treatment of PC and as siRNAs are currently being applied in clinical trials, significant progress can be made by translating the current findings into the clinical settings.
Collapse
|
72
|
Bofinger R, Weitsman G, Evans R, Glaser M, Sander K, Allan H, Hochhauser D, Kalber TL, Årstad E, Hailes HC, Ng T, Tabor AB. Drug delivery, biodistribution and anti-EGFR activity: theragnostic nanoparticles for simultaneous in vivo delivery of tyrosine kinase inhibitors and kinase activity biosensors. NANOSCALE 2021; 13:18520-18535. [PMID: 34730152 PMCID: PMC8601123 DOI: 10.1039/d1nr02770k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 09/24/2021] [Indexed: 05/03/2023]
Abstract
In vivo delivery of small molecule therapeutics to cancer cells, assessment of the selectivity of administration, and measuring the efficacity of the drug in question at the molecule level, are important ongoing challenges in developing new classes of cancer chemotherapeutics. One approach that has the potential to provide targeted delivery, tracking of biodistribution and readout of efficacy, is to use multimodal theragnostic nanoparticles to deliver the small molecule therapeutic. In this paper, we report the development of targeted theragnostic lipid/peptide/DNA lipopolyplexes. These simultaneously deliver an inhibitor of the EGFR tyrosine kinase, and plasmid DNA coding for a Crk-based biosensor, Picchu-X, which when expressed in the target cells can be used to quantify the inhibition of EGFR in vivo in a mouse colorectal cancer xenograft model. Reversible bioconjugation of a known analogue of the tyrosine kinase inhibitor Mo-IPQA to a cationic peptide, and co-formulation with peptides containing both EGFR-binding and cationic sequences, allowed for good levels of inhibitor encapsulation with targeted delivery to LIM1215 colon cancer cells. Furthermore, high levels of expression of the Picchu-X biosensor in the LIM1215 cells in vivo allowed us to demonstrate, using fluorescence lifetime microscopy (FLIM)-based biosensing, that EGFR activity can be successfully suppressed by the tyrosine kinase inhibitor, released from the lipopolyplexes. Finally, we measured the biodistribution of lipopolyplexes containing 125I-labelled inhibitors and were able to demonstrate that the lipopolyplexes gave significantly higher drug delivery to the tumors compared with free drug.
Collapse
Affiliation(s)
- Robin Bofinger
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
| | - Gregory Weitsman
- School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK.
| | - Rachel Evans
- School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK.
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Matthias Glaser
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
- Centre for Radiopharmaceutical Chemistry, Kathleen Lonsdale Building, 5 Gower Place, London WC1E 6BS, UK
| | - Kerstin Sander
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
- Centre for Radiopharmaceutical Chemistry, Kathleen Lonsdale Building, 5 Gower Place, London WC1E 6BS, UK
| | - Helen Allan
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
| | - Daniel Hochhauser
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Erik Årstad
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
- Centre for Radiopharmaceutical Chemistry, Kathleen Lonsdale Building, 5 Gower Place, London WC1E 6BS, UK
| | - Helen C Hailes
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
| | - Tony Ng
- School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK.
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Alethea B Tabor
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
| |
Collapse
|
73
|
Novel Peptide Therapeutic Approaches for Cancer Treatment. Cells 2021; 10:cells10112908. [PMID: 34831131 PMCID: PMC8616177 DOI: 10.3390/cells10112908] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 11/17/2022] Open
Abstract
Peptides are increasingly being developed for use as therapeutics to treat many ailments, including cancer. Therapeutic peptides have the advantages of target specificity and low toxicity. The anticancer effects of a peptide can be the direct result of the peptide binding its intended target, or the peptide may be conjugated to a chemotherapy drug or radionuclide and used to target the agent to cancer cells. Peptides can be targeted to proteins on the cell surface, where the peptide–protein interaction can initiate internalization of the complex, or the peptide can be designed to directly cross the cell membrane. Peptides can induce cell death by numerous mechanisms including membrane disruption and subsequent necrosis, apoptosis, tumor angiogenesis inhibition, immune regulation, disruption of cell signaling pathways, cell cycle regulation, DNA repair pathways, or cell death pathways. Although using peptides as therapeutics has many advantages, peptides have the disadvantage of being easily degraded by proteases once administered and, depending on the mode of administration, often have difficulty being adsorbed into the blood stream. In this review, we discuss strategies recently developed to overcome these obstacles of peptide delivery and bioavailability. In addition, we present many examples of peptides developed to fight cancer.
Collapse
|
74
|
Chimeric virus-like particles (VLPs) designed from shrimp nodavirus (MrNV) capsid protein specifically target EGFR-positive human colorectal cancer cells. Sci Rep 2021; 11:16579. [PMID: 34400669 PMCID: PMC8367941 DOI: 10.1038/s41598-021-95891-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 07/30/2021] [Indexed: 12/24/2022] Open
Abstract
Recombinant MrNV capsid protein has been shown to effectively deliver plasmid DNA and dsRNA into Sf9 insect cells and shrimp tissues. To extend its application to cancer cell-targeting drug delivery, we created three different types of chimeric MrNV virus-like particles (VLPs) (R-MrNV, I-MrNV, and E-MrNV) that have specificity toward the epidermal growth factor receptor (EGFR), a cancer cell biomarker, by incorporating the EGFR-specific GE11 peptide at 3 different locations within the host cell recognition site of the capsid. All three chimeric MrNV-VLPs preserved the ability to form a mulberry-like VLP structure and to encapsulate EGFP DNA plasmid with an efficiency comparable to that previously reported for normal MrNV (N-MrNV). Compared to N-MrNV, the chimeric R-MrNV and E-MrNV carrying the exposed GE-11 peptide showed a significantly enhanced binding and internalization abilities that were specific towards EGFR expression in colorectal cancer cells (SW480). Specific targeting of chimeric MrNV to EGFR was proven by both EGFR silencing with siRNA vector and a competition with excess GE-11 peptide as well as the use of EGFR-negative colorectal cells (SW620) and breast cancer cells (MCF7). We demonstrated here that both chimeric R-MrNV and E-MrNV could be used to encapsulate cargo such as exogenous DNA and deliver it specifically to EGFR-positive cells. Our study presents the potential use of surface-modified VLPs of shrimp virus origin as nanocontainers for targeted cancer drug delivery.
Collapse
|
75
|
Zheng M, Li C, Zhou M, Jia R, Cai G, She F, Wei L, Wang S, Yu J, Wang D, Calcul L, Sun X, Luo X, Cheng F, Li Q, Wang Y, Cai J. Discovery of Cyclic Peptidomimetic Ligands Targeting the Extracellular Domain of EGFR. J Med Chem 2021; 64:11219-11228. [PMID: 34297567 DOI: 10.1021/acs.jmedchem.1c00607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
It is very promising to target the extracellular domain of epidermal growth factor receptor (EGFR) for developing novel and selective anticancer therapies. Herein, we report the discovery of a novel small molecule, M-2-5, from a one-bead-two-compound (OBTC) cyclic γ-AApeptide library. The molecule was found to bind tightly to the extracellular domain of EGFR. Intriguingly, this molecule could also effectively antagonize EGF-stimulated EGFR phosphorylation and downstream signal transduction. Furthermore, together with its remarkable resistance to proteolytic degradation, M-2-5 was shown to effectively inhibit cell proliferation and migration in vitro and suppresses the growth of tumor in the A549 xenograft model in vivo, highlighting its potential therapeutic application for cancer treatment.
Collapse
Affiliation(s)
- Mengmeng Zheng
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, Florida 33620, United States
| | - Chunpu Li
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, Florida 33620, United States.,Department of Medical Oncology & Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mi Zhou
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, Florida 33620, United States
| | - Ru Jia
- Department of Medical Oncology & Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Gang Cai
- Department of Medical Oncology & Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fengyu She
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, Florida 33620, United States
| | - Lulu Wei
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, Florida 33620, United States
| | - Shaohui Wang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Jie Yu
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Dingyan Wang
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Laurent Calcul
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, Florida 33620, United States
| | - Xingmin Sun
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Xiaomin Luo
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Feng Cheng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida 33612, United States
| | - Qi Li
- Department of Medical Oncology & Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yan Wang
- Department of Medical Oncology & Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, Florida 33620, United States
| |
Collapse
|
76
|
Abstract
Breast cancer continues to be the most lethal cancer type in women and one of the most diagnosed. Understanding Breast cancer receptor status is one of the most vital processes for determining treatment options. One type of breast cancer, human epidermal growth factor receptor 2 (HER2) positive, has approved receptor-based therapies including trastuzumab and pertuzumab that can significantly increase the likelihood of survival. Current methods to determine HER2 status include biopsies with immunohistochemical staining and/or fluorescence in situ hybridization. However, positron emission tomography (PET) imaging techniques using 89Zr-trastuzumab or 89Zr-pertuzumab are currently in clinical trials for a non-invasive, full body diagnostic approach. Although the antibodies have strong specificity to the HER2 positive lesions, challenges involving long post-injection time for imaging due to the blood circulation of the antibodies and matching of long-live isotopes leading to increased dose to the patient leave opportunities for alternative PET imaging probes. Peptides have been shown to allow for shorter injection-to-imaging time and can be used with shorter lived isotopes. HER2 specific peptides under development will help improve the diagnosis and potentially therapy options for HER2 positive breast cancer. Peptides showing specificity for HER2 could start widespread development of molecular imaging techniques for HER2 positive cancers.
Collapse
Affiliation(s)
- Maxwell Ducharme
- Department of Radiology, 9968University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suzanne E Lapi
- Department of Radiology, 9968University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
77
|
Liu M, Fang X, Yang Y, Wang C. Peptide-Enabled Targeted Delivery Systems for Therapeutic Applications. Front Bioeng Biotechnol 2021; 9:701504. [PMID: 34277592 PMCID: PMC8281044 DOI: 10.3389/fbioe.2021.701504] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Receptor-targeting peptides have been extensively pursued for improving binding specificity and effective accumulation of drugs at the site of interest, and have remained challenging for extensive research efforts relating to chemotherapy in cancer treatments. By chemically linking a ligand of interest to drug-loaded nanocarriers, active targeting systems could be constructed. Peptide-functionalized nanostructures have been extensively pursued for biomedical applications, including drug delivery, biological imaging, liquid biopsy, and targeted therapies, and widely recognized as candidates of novel therapeutics due to their high specificity, well biocompatibility, and easy availability. We will endeavor to review a variety of strategies that have been demonstrated for improving receptor-specificity of the drug-loaded nanoscale structures using peptide ligands targeting tumor-related receptors. The effort could illustrate that the synergism of nano-sized structures with receptor-targeting peptides could lead to enrichment of biofunctions of nanostructures.
Collapse
Affiliation(s)
- Mingpeng Liu
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Department of Chemistry, Tsinghua University, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaocui Fang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yanlian Yang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chen Wang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
78
|
Biscaglia F, Caligiuri I, Rizzolio F, Ripani G, Palleschi A, Meneghetti M, Gobbo M. Protection against proteolysis of a targeting peptide on gold nanostructures. NANOSCALE 2021; 13:10544-10554. [PMID: 34100487 DOI: 10.1039/d0nr04631k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cell targeting has been considered an important strategy in diagnostic and therapeutic applications. Among different targeting units, peptides have emerged for their ability to bind to many different cellular targets, their scarce immunogenicity and the possibility of introducing multiple copies on nanosystems, providing high avidity for the target. However, their sensitivity to proteases strongly limits their applications in vivo. Here, we show that when presented on the surface of nanostructures, peptide stability to proteolysis is strongly improved without reducing the targeting activity. We prepared plasmonic nanostructures functionalized with a dodecapeptide (GE11) which targets EGFR, a protein overexpressed on different types of tumors. Two types of nanosystems were prepared in which the targeting unit was either directly linked to gold nanoparticles or through a PEG chain, resulting in a different peptide density on the surface of nanostructures. The peptide was rapidly degraded in 20% human serum or in the presence of isolated serine proteases, whereas no significant proteolytic fragments were detected during incubation of the nanosystems and after 24 h digestion, the nanostructures maintained their targeting activity and selectivity on colon cancer cells. Molecular dynamic calculations of the interaction of the nanostructure with chymotrypsin suggest that the formation of the enzyme-peptide complex, the first step in the mechanism of peptide hydrolysis, is highly unlikely because of the constraint imposed by the link of the peptide to the nanoparticle. These results support the utilization of peptides as active targeting units in nanomedicine.
Collapse
Affiliation(s)
- Francesca Biscaglia
- Department of Chemical Sciences, University of Padova, via F. Marzolo 1, 35131 Padova, Italy.
| | | | | | | | | | | | | |
Collapse
|
79
|
Nanomedicines functionalized with anti-EGFR ligands for active targeting in cancer therapy: Biological strategy, design and quality control. Int J Pharm 2021; 605:120795. [PMID: 34119579 DOI: 10.1016/j.ijpharm.2021.120795] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
Recently, active targeting using nanocarriers with biological ligands has emerged as a novel strategy for improving the delivery of therapeutic and/or imaging agents to tumor cells. The presence of active targeting moieties on the surface of nanomedicines has been shown to play an important role in enhancing their accumulation in tumoral cells and tissues versus healthy ones. This property not only helps to increase the therapeutic index but also to minimize possible side effects of the designed nanocarriers. Since the overexpression of epidermal growth factor receptors (EGFR) is a common occurrence linked to the progression of a broad variety of cancers, the potential application of anti-EGFR immunotherapy and EGFR-targeting ligands in active targeting nanomedicines is getting increasing attention. Henceforth, the EGFR-targeted nanomedicines were extensively studied in vitro and in vivo but exhibited both satisfactory and disappointing results, depending on used protocols. This review is designed to give an overview of a variety of EGFR-targeting ligands available for nanomedicines, how to conjugate them onto the surface of nanoparticles, and the main analytical methods to confirm this successful conjugation.
Collapse
|
80
|
Li K, Pang L, Pan X, Fan S, Wang X, Wang Q, Dai P, Gao W, Gao J. GE11 Modified PLGA/TPGS Nanoparticles Targeting Delivery of Salinomycin to Breast Cancer Cells. Technol Cancer Res Treat 2021; 20:15330338211004954. [PMID: 34056977 PMCID: PMC8182624 DOI: 10.1177/15330338211004954] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Salinomycin (Sal) is a potent inhibitor with effective anti-breast cancer properties in clinical therapy. The occurrence of various side effect of Sal greatly limits its application. The epidermal growth factor receptor (EGFR) family is a family of receptors highly expressed in most breast cancer cells. GE11 is a dodecapeptide which shows excellent EGFR affinity. A series of nanoparticles derivatives with GE11 peptide conjugated PLGA/TPGS were synthesized. Nanoprecipitation method was used to prepare the Sal loaded nanoparticles at the optimized concentration. The characterization, targeting efficacy, and antitumor activity were detected both in vitro and in vivo. Encapsulation of Sal in GE11 modified PLGA/TPGS nanoparticles shows an improved therapy efficacy and lower systemic side effect. This represents the delivery system a promising strategy to enhance the therapeutic effect against EGFR highly expressed breast cancer.
Collapse
Affiliation(s)
- Kaichun Li
- Department of Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Liying Pang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xiaorong Pan
- Department of Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Shaonan Fan
- Department of Radiation Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Xinxin Wang
- Department of Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Qiaoyun Wang
- Department of Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Ping Dai
- Department of Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Wei Gao
- Department of Radiation Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Jie Gao
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
81
|
Cao XH, Liang MX, Wu Y, Yang K, Tang JH, Zhang W. Extracellular vesicles as drug vectors for precise cancer treatment. Nanomedicine (Lond) 2021; 16:1519-1537. [PMID: 34011162 DOI: 10.2217/nnm-2021-0123] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are nano-sized vesicle structures secreted from a variety of cells, which carry numerous biological macromolecules, participate in cell signal transduction and avoid immune system clearance. EVs have a plethora of specific signal recognition factors, and many studies have shown that they can play an important role in the precise treatment of tumors. This review aims to compile the applications of EVs as nanocarriers for antitumor drugs, gene drugs and other nanomaterials with anticancer capability. Additionally, we systematically summarize the preparation methodology and expound upon how to improve the drug loading and cancer-targeting capacity of EVs. We highlight that EV-based drug delivery has the potential to become the future of precise cancer treatment.
Collapse
Affiliation(s)
- Xin-Hui Cao
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China.,School of Clinical Medicine, Xuzhou Medical University, Xuzhou 221000, PR China
| | - Ming-Xing Liang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Yang Wu
- Biobank, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Kai Yang
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou 221000, PR China
| | - Jin-Hai Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China.,School of Clinical Medicine, Xuzhou Medical University, Xuzhou 221000, PR China
| | - Wei Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| |
Collapse
|
82
|
Geskovski N, Matevska-Geshkovska N, Dimchevska Sazdovska S, Glavas Dodov M, Mladenovska K, Goracinova K. The impact of molecular tumor profiling on the design strategies for targeting myeloid leukemia and EGFR/CD44-positive solid tumors. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:375-401. [PMID: 33981532 PMCID: PMC8093552 DOI: 10.3762/bjnano.12.31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/12/2021] [Indexed: 05/21/2023]
Abstract
Nanomedicine has emerged as a novel cancer treatment and diagnostic modality, whose design constantly evolves towards increasing the safety and efficacy of the chemotherapeutic and diagnostic protocols. Molecular diagnostics, which create a great amount of data related to the unique molecular signatures of each tumor subtype, have emerged as an important tool for detailed profiling of tumors. They provide an opportunity to develop targeting agents for early detection and diagnosis, and to select the most effective combinatorial treatment options. Alongside, the design of the nanoscale carriers needs to cope with novel trends of molecular screening. Also, multiple targeting ligands needed for robust and specific interactions with the targeted cell populations have to be introduced, which should result in substantial improvements in safety and efficacy of the cancer treatment. This article will focus on novel design strategies for nanoscale drug delivery systems, based on the unique molecular signatures of myeloid leukemia and EGFR/CD44-positive solid tumors, and the impact of novel discoveries in molecular tumor profiles on future chemotherapeutic protocols.
Collapse
Affiliation(s)
- Nikola Geskovski
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Nadica Matevska-Geshkovska
- Center for Pharmaceutical Biomolecular Analyses, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Simona Dimchevska Sazdovska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
- Department of Nanobiotechnology, Institute of Biotechnology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Marija Glavas Dodov
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Kristina Mladenovska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Katerina Goracinova
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
- College of Pharmacy, Qatar University, PO Box 2713, Doha, Qatar
| |
Collapse
|
83
|
Aloisio A, Nisticò N, Mimmi S, Maisano D, Vecchio E, Fiume G, Iaccino E, Quinto I. Phage-Displayed Peptides for Targeting Tyrosine Kinase Membrane Receptors in Cancer Therapy. Viruses 2021; 13:649. [PMID: 33918836 PMCID: PMC8070105 DOI: 10.3390/v13040649] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) regulate critical physiological processes, such as cell growth, survival, motility, and metabolism. Abnormal activation of RTKs and relative downstream signaling is implicated in cancer pathogenesis. Phage display allows the rapid selection of peptide ligands of membrane receptors. These peptides can target in vitro and in vivo tumor cells and represent a novel therapeutic approach for cancer therapy. Further, they are more convenient compared to antibodies, being less expensive and non-immunogenic. In this review, we describe the state-of-the-art of phage display for development of peptide ligands of tyrosine kinase membrane receptors and discuss their potential applications for tumor-targeted therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ileana Quinto
- Correspondence: (A.A.); (I.Q.): Tel.: +39-0961-3694057 (I.Q.)
| |
Collapse
|
84
|
Ayo A, Laakkonen P. Peptide-Based Strategies for Targeted Tumor Treatment and Imaging. Pharmaceutics 2021; 13:pharmaceutics13040481. [PMID: 33918106 PMCID: PMC8065807 DOI: 10.3390/pharmaceutics13040481] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/03/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide. The development of cancer-specific diagnostic agents and anticancer toxins would improve patient survival. The current and standard types of medical care for cancer patients, including surgery, radiotherapy, and chemotherapy, are not able to treat all cancers. A new treatment strategy utilizing tumor targeting peptides to selectively deliver drugs or applicable active agents to solid tumors is becoming a promising approach. In this review, we discuss the different tumor-homing peptides discovered through combinatorial library screening, as well as native active peptides. The different structure–function relationship data that have been used to improve the peptide’s activity and conjugation strategies are highlighted.
Collapse
Affiliation(s)
- Abiodun Ayo
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Laboratory Animal Center, HiLIFE—Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-50-4489100
| |
Collapse
|
85
|
Rinne SS, Orlova A, Tolmachev V. PET and SPECT Imaging of the EGFR Family (RTK Class I) in Oncology. Int J Mol Sci 2021; 22:ijms22073663. [PMID: 33915894 PMCID: PMC8036874 DOI: 10.3390/ijms22073663] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
The human epidermal growth factor receptor family (EGFR-family, other designations: HER family, RTK Class I) is strongly linked to oncogenic transformation. Its members are frequently overexpressed in cancer and have become attractive targets for cancer therapy. To ensure effective patient care, potential responders to HER-targeted therapy need to be identified. Radionuclide molecular imaging can be a key asset for the detection of overexpression of EGFR-family members. It meets the need for repeatable whole-body assessment of the molecular disease profile, solving problems of heterogeneity and expression alterations over time. Tracer development is a multifactorial process. The optimal tracer design depends on the application and the particular challenges of the molecular target (target expression in tumors, endogenous expression in healthy tissue, accessibility). We have herein summarized the recent preclinical and clinical data on agents for Positron Emission Tomography (PET) and Single Photon Emission Tomography (SPECT) imaging of EGFR-family receptors in oncology. Antibody-based tracers are still extensively investigated. However, their dominance starts to be challenged by a number of tracers based on different classes of targeting proteins. Among these, engineered scaffold proteins (ESP) and single domain antibodies (sdAb) show highly encouraging results in clinical studies marking a noticeable trend towards the use of smaller sized agents for HER imaging.
Collapse
Affiliation(s)
- Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden; (S.S.R.); (A.O.)
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden; (S.S.R.); (A.O.)
- Science for Life Laboratory, Uppsala University, 752 37 Uppsala, Sweden
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Vladimir Tolmachev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 37 Uppsala, Sweden
- Correspondence: ; Tel.: +46-704-250-782
| |
Collapse
|
86
|
Ahmad E, Ali A, Fatima MT, Nimisha, Apurva, Kumar A, Sumi MP, Sattar RSA, Mahajan B, Saluja SS. Ligand decorated biodegradable nanomedicine in the treatment of cancer. Pharmacol Res 2021; 167:105544. [PMID: 33722711 DOI: 10.1016/j.phrs.2021.105544] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 12/14/2022]
Abstract
Cancer is one of the major global health problems, responsible for the second-highest number of deaths. The genetic and epigenetic changes in the oncogenes or tumor suppressor genes alter the regulatory pathways leading to its onset and progression. Conventional methods are used in appropriate combinations for the treatment. Surgery effectively treats localized tumors; however, it fails to treat metastatic tumors, leading to a spread in other organs, causing a high recurrence rate and death. Among the different strategies, the nanocarriers-based approach is highly sought for, but its nonspecific delivery can cause a profound side effect on healthy cells. Targeted nanomedicine has the advantage of targeting cancer cells specifically by interacting with the receptors overexpressed on their surface, overcoming its non-specificity to target healthy cells. Nanocarriers prepared from biodegradable and biocompatible materials are decorated with different ligands by encapsulating therapeutic or diagnostic agents or both to target cancer cells overexpressing the receptors. Scientists are now utilizing a theranostic approach to simultaneously evaluate nanocarrier bio-distribution and its effect on the treatment regime. Herein, we have summarized the recent 5-year efforts in the development of the ligands decorated biodegradable nanocarriers, as a targeted nanomedicine approach, which has been highly promising in the treatment of cancer.
Collapse
Affiliation(s)
- Ejaj Ahmad
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Asgar Ali
- Department of Biochemistry, All India Institute of Medical Science, Patna 810507, India
| | - Munazza Tamkeen Fatima
- Department of Pharmaceutical Science, College of Pharmacy, QU health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Apurva
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Arun Kumar
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Mamta P Sumi
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Real Sumayya Abdul Sattar
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India
| | - Bhawna Mahajan
- Department of Biochemistry, Govind Ballabh Pant, Postgraduate Institute of Medical, Education and Research (GIPMER), New Delhi 110002, India
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India; Department of GI Surgery, Govind Ballabh Pant, Postgraduate Institute of Medica, Education and Research (GIPMER), New Delhi 110002, India.
| |
Collapse
|
87
|
Aronson MR, Medina SH, Mitchell MJ. Peptide functionalized liposomes for receptor targeted cancer therapy. APL Bioeng 2021; 5:011501. [PMID: 33532673 PMCID: PMC7837755 DOI: 10.1063/5.0029860] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023] Open
Abstract
Most clinically approved cancer therapies are potent and toxic small molecules that are limited by severe off-target toxicities and poor tumor-specific localization. Over the past few decades, attempts have been made to load chemotherapies into liposomes, which act to deliver the therapeutic agent directly to the tumor. Although liposomal encapsulation has been shown to decrease toxicity in human patients, reliance on passive targeting via the enhanced permeability and retention (EPR) effect has left some of these issues unresolved. Recently, investigations into modifying the surface of liposomes via covalent and/or electrostatic functionalization have offered mechanisms for tumor homing and subsequently controlled chemotherapeutic delivery. A wide variety of biomolecules can be utilized to functionalize liposomes such as proteins, carbohydrates, and nucleic acids, which enable multiple directions for cancer cell localization. Importantly, when nanoparticles are modified with such molecules, care must be taken as not to inactivate or denature the ligand. Peptides, which are small proteins with <30 amino acids, have demonstrated the exceptional ability to act as ligands for transmembrane protein receptors overexpressed in many tumor phenotypes. Exploring this strategy offers a method in tumor targeting for cancers such as glioblastoma multiforme, pancreatic, lung, and breast based on the manifold of receptors overexpressed on various tumor cell populations. In this review, we offer a comprehensive summary of peptide-functionalized liposomes for receptor-targeted cancer therapy.
Collapse
|
88
|
Molecular Targeting of Epidermal Growth Factor Receptor (EGFR) and Vascular Endothelial Growth Factor Receptor (VEGFR). Molecules 2021; 26:molecules26041076. [PMID: 33670650 PMCID: PMC7922143 DOI: 10.3390/molecules26041076] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/13/2021] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) are two extensively studied membrane-bound receptor tyrosine kinase proteins that are frequently overexpressed in many cancers. As a result, these receptor families constitute attractive targets for imaging and therapeutic applications in the detection and treatment of cancer. This review explores the dynamic structure and structure-function relationships of these two growth factor receptors and their significance as it relates to theranostics of cancer, followed by some of the common inhibition modalities frequently employed to target EGFR and VEGFR, such as tyrosine kinase inhibitors (TKIs), antibodies, nanobodies, and peptides. A summary of the recent advances in molecular imaging techniques, including positron emission tomography (PET), single-photon emission computerized tomography (SPECT), computed tomography (CT), magnetic resonance imaging (MRI), and optical imaging (OI), and in particular, near-IR fluorescence imaging using tetrapyrrolic-based fluorophores, concludes this review.
Collapse
|
89
|
Jayasinghe MK, Tan M, Peng B, Yang Y, Sethi G, Pirisinu M, Le MTN. New approaches in extracellular vesicle engineering for improving the efficacy of anti-cancer therapies. Semin Cancer Biol 2021; 74:62-78. [PMID: 33609665 DOI: 10.1016/j.semcancer.2021.02.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/11/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Cancer is a disease that evolves continuously with unpredictable outcomes. Although conventional chemotherapy can display significant antitumor effects, the lack of specificity and poor bioavailability remain major concerns in cancer therapy. Moreover, with the advent of novel anti-cancer gene therapies, there is an urgent need for drug delivery vectors capable of bypassing cellular barriers and efficiently transferring therapeutic cargo to recipient cells. A number of drug delivery systems have been proposed to overcome these limitations, but their successful clinical translation has been hampered by the onset of unexpected side effects and associated toxicities. The application of extracellular vesicles (EVs), a class of naturally released, cell-derived particles, as drug delivery vectors presents a breakthrough in nanomedicine, taking into account their biocompatibility and natural role in intercellular communication. Combining the advantageous intrinsic properties of EVs with surface functionalization and the encapsulation of drugs allows for a new class of engineered EVs that serve as effective therapeutic carriers. Here, we describe the various successful approaches involving the application of engineered EVs as bio-derived drug delivery vectors in cancer therapy. The latest and most effective strategies of engineering EVs to improve drug loading, stealth properties and tumour targeting capabilities of EVs are debated. Finally, current obstacles and future perspectives of smart engineered EVs are discussed.
Collapse
Affiliation(s)
- Migara Kavishka Jayasinghe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Digital Medicine, Immunology Programme and Cancer Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore; Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong
| | - Melissa Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Digital Medicine, Immunology Programme and Cancer Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Boya Peng
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Digital Medicine, Immunology Programme and Cancer Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Yuqi Yang
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Marco Pirisinu
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong.
| | - Minh T N Le
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Digital Medicine, Immunology Programme and Cancer Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
90
|
Pham TC, Jayasinghe MK, Pham TT, Yang Y, Wei L, Usman WM, Chen H, Pirisinu M, Gong J, Kim S, Peng B, Wang W, Chan C, Ma V, Nguyen NT, Kappei D, Nguyen X, Cho WC, Shi J, Le MT. Covalent conjugation of extracellular vesicles with peptides and nanobodies for targeted therapeutic delivery. J Extracell Vesicles 2021; 10:e12057. [PMID: 33643546 PMCID: PMC7886705 DOI: 10.1002/jev2.12057] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 10/16/2020] [Accepted: 12/14/2020] [Indexed: 12/15/2022] Open
Abstract
Natural extracellular vesicles (EVs) are ideal drug carriers due to their remarkable biocompatibility. Their delivery specificity can be achieved by the conjugation of targeting ligands. However, existing methods to engineer target-specific EVs are tedious or inefficient, having to compromise between harsh chemical treatments and transient interactions. Here, we describe a novel method for the covalent conjugation of EVs with high copy numbers of targeting moieties using protein ligases. Conjugation of EVs with either an epidermal growth factor receptor (EGFR)-targeting peptide or anti-EGFR nanobody facilitates their accumulation in EGFR-positive cancer cells, both in vitro and in vivo. Systemic delivery of paclitaxel by EGFR-targeting EVs at a low dose significantly increases drug efficacy in a xenografted mouse model of EGFR-positive lung cancer. The method is also applicable to the conjugation of EVs with peptides and nanobodies targeting other receptors, such as HER2 and SIRP alpha, and the conjugated EVs can deliver RNA in addition to small molecules, supporting the versatile application of EVs in cancer therapies. This simple, yet efficient and versatile method for the stable surface modification of EVs bypasses the need for genetic and chemical modifications, thus facilitating safe and specific delivery of therapeutic payloads to target cells.
Collapse
Affiliation(s)
- Tin Chanh Pham
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
| | - Migara Kavishka Jayasinghe
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
- Institute for Digital MedicineImmunology Programme and Cancer ProgrammeYong Loo Lin School of MedicineNational University of SingaporeSingapore
- N.1 Institute for HealthNational University of SingaporeSingapore
| | - Thach Tuan Pham
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
- Institute for Digital MedicineImmunology Programme and Cancer ProgrammeYong Loo Lin School of MedicineNational University of SingaporeSingapore
- N.1 Institute for HealthNational University of SingaporeSingapore
| | - Yuqi Yang
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
| | - Likun Wei
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
- City University of Hong Kong Shenzhen InstituteShenzhenChina
| | - Waqas Muhammad Usman
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
| | - Huan Chen
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
| | - Marco Pirisinu
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
| | - Jinhua Gong
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
- City University of Hong Kong Shenzhen InstituteShenzhenChina
| | - Seongkyeol Kim
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
| | - Boya Peng
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore
- Institute for Digital MedicineImmunology Programme and Cancer ProgrammeYong Loo Lin School of MedicineNational University of SingaporeSingapore
- N.1 Institute for HealthNational University of SingaporeSingapore
| | - Weixi Wang
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
- City University of Hong Kong Shenzhen InstituteShenzhenChina
| | - Charlene Chan
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
| | - Victor Ma
- Department of Clinical OncologyQueen Elizabeth HospitalHong Kong
| | - Nhung T.H. Nguyen
- Vinmec Institute of Applied Science and Regenerative Medicineand College of Health SciencesVinmec Healthcare systemVin UniversityHanoiVietnam
| | - Dennis Kappei
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
- Department of BiochemistryYong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Xuan‐Hung Nguyen
- Vinmec Institute of Applied Science and Regenerative Medicineand College of Health SciencesVinmec Healthcare systemVin UniversityHanoiVietnam
| | - William C. Cho
- Department of Clinical OncologyQueen Elizabeth HospitalHong Kong
| | - Jiahai Shi
- Department of Biomedical SciencesCollege of Veterinary Medicine and Life SciencesCity University of Hong KongHong Kong
- City University of Hong Kong Shenzhen InstituteShenzhenChina
| | - Minh T.N. Le
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore
- Institute for Digital MedicineImmunology Programme and Cancer ProgrammeYong Loo Lin School of MedicineNational University of SingaporeSingapore
- N.1 Institute for HealthNational University of SingaporeSingapore
| |
Collapse
|
91
|
Williams TM, Kaufman NEM, Zhou Z, Singh SS, Jois SD, Vicente MDGH. Click Conjugation of Boron Dipyrromethene (BODIPY) Fluorophores to EGFR-Targeting Linear and Cyclic Peptides. Molecules 2021; 26:molecules26030593. [PMID: 33498632 PMCID: PMC7865655 DOI: 10.3390/molecules26030593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 12/15/2022] Open
Abstract
Through a simple 1,3-cycloaddition reaction, three BODIPY-peptide conjugates that target the extracellular domain of the epidermal growth factor receptor (EGFR) were prepared and their ability for binding to EGFR was investigated. The peptide ligands K(N3)LARLLT and its cyclic analog cyclo(K(N3)larllt, previously shown to have high affinity for binding to the extracellular domain of EGFR, were conjugated to alkynyl-functionalized BODIPY dyes 1 and 2 via a copper-catalyzed click reaction. This reaction produced conjugates 3, 4, and 5 in high yields (70–82%). In vitro studies using human carcinoma HEp2 cells that overexpress EGFR demonstrated high cellular uptake, particularly for the cyclic peptide conjugate 5, and low cytotoxicity in light (~1 J·cm−2) and darkness. Surface plasmon resonance (SPR) results show binding affinity of the three BODIPY-peptide conjugates for EGFR, particularly for 5 bearing the cyclic peptide. Competitive binding studies using three cell lines with different expressions of EGFR show that 5 binds specifically to EGFR-overexpressing colon cancer cells. Among the three conjugates, 5 bearing the cyclic peptide exhibited the highest affinity for binding to the EGFR protein.
Collapse
Affiliation(s)
- Tyrslai M. Williams
- Department of Chemistry, Louisiana State University, Baton Rouge, LA 70803, USA; (T.M.W.); (N.E.M.K.); (Z.Z.)
| | - Nichole E. M. Kaufman
- Department of Chemistry, Louisiana State University, Baton Rouge, LA 70803, USA; (T.M.W.); (N.E.M.K.); (Z.Z.)
| | - Zehua Zhou
- Department of Chemistry, Louisiana State University, Baton Rouge, LA 70803, USA; (T.M.W.); (N.E.M.K.); (Z.Z.)
| | - Sitanshu S. Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA; (S.S.S.); (S.D.J.)
| | - Seetharama D. Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA; (S.S.S.); (S.D.J.)
| | - Maria da Graça H. Vicente
- Department of Chemistry, Louisiana State University, Baton Rouge, LA 70803, USA; (T.M.W.); (N.E.M.K.); (Z.Z.)
- Correspondence: ; Tel.: +1-225-578-7405; Fax: +1-225-578-3458
| |
Collapse
|
92
|
Tang SY, Wei H, Yu CY. Peptide-functionalized delivery vehicles for enhanced cancer therapy. Int J Pharm 2021; 593:120141. [DOI: 10.1016/j.ijpharm.2020.120141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/17/2020] [Accepted: 11/28/2020] [Indexed: 02/08/2023]
|
93
|
EGFR-Binding Peptides: From Computational Design towards Tumor-Targeting of Adeno-Associated Virus Capsids. Int J Mol Sci 2020; 21:ijms21249535. [PMID: 33333826 PMCID: PMC7765298 DOI: 10.3390/ijms21249535] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/04/2020] [Accepted: 12/06/2020] [Indexed: 01/24/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) plays a central role in the progression of many solid tumors. We used this validated target to analyze the de novo design of EGFR-binding peptides and their application for the delivery of complex payloads via rational design of a viral vector. Peptides were computationally designed to interact with the EGFR dimerization interface. Two new peptides and a reference (EDA peptide) were chemically synthesized, and their binding ability characterized. Presentation of these peptides in each of the 60 capsid proteins of recombinant adeno-associated viruses (rAAV) via a genetic based loop insertion enabled targeting of EGFR overexpressing tumor cell lines. Furthermore, tissue distribution and tumor xenograft specificity were analyzed with systemic injection in chicken egg chorioallantoic membrane (CAM) assays. Complex correlations between the targeting of the synthetic peptides and the viral vectors to cells and in ovo were observed. Overall, these data demonstrate the potential of computational design in combination with rational capsid modification for viral vector targeting opening new avenues for viral vector delivery and specifically suicide gene therapy.
Collapse
|
94
|
Gierlich P, Mata AI, Donohoe C, Brito RMM, Senge MO, Gomes-da-Silva LC. Ligand-Targeted Delivery of Photosensitizers for Cancer Treatment. Molecules 2020; 25:E5317. [PMID: 33202648 PMCID: PMC7698280 DOI: 10.3390/molecules25225317] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/26/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising cancer treatment which involves a photosensitizer (PS), light at a specific wavelength for PS activation and oxygen, which combine to elicit cell death. While the illumination required to activate a PS imparts a certain amount of selectivity to PDT treatments, poor tumor accumulation and cell internalization are still inherent properties of most intravenously administered PSs. As a result, common consequences of PDT include skin photosensitivity. To overcome the mentioned issues, PSs may be tailored to specifically target overexpressed biomarkers of tumors. This active targeting can be achieved by direct conjugation of the PS to a ligand with enhanced affinity for a target overexpressed on cancer cells and/or other cells of the tumor microenvironment. Alternatively, PSs may be incorporated into ligand-targeted nanocarriers, which may also encompass multi-functionalities, including diagnosis and therapy. In this review, we highlight the major advances in active targeting of PSs, either by means of ligand-derived bioconjugates or by exploiting ligand-targeting nanocarriers.
Collapse
Affiliation(s)
- Piotr Gierlich
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Ana I. Mata
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
| | - Claire Donohoe
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Rui M. M. Brito
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- BSIM Therapeutics, Instituto Pedro Nunes, 3030-199 Coimbra, Portugal
| | - Mathias O. Senge
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Lígia C. Gomes-da-Silva
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
| |
Collapse
|
95
|
Cunliffe TG, Bates EA, Parker AL. Hitting the Target but Missing the Point: Recent Progress towards Adenovirus-Based Precision Virotherapies. Cancers (Basel) 2020; 12:E3327. [PMID: 33187160 PMCID: PMC7696810 DOI: 10.3390/cancers12113327] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/31/2020] [Accepted: 11/09/2020] [Indexed: 12/23/2022] Open
Abstract
More people are surviving longer with cancer. Whilst this can be partially attributed to advances in early detection of cancers, there is little doubt that the improvement in survival statistics is also due to the expansion in the spectrum of treatments available for efficacious treatment. Transformative amongst those are immunotherapies, which have proven effective agents for treating immunogenic forms of cancer, although immunologically "cold" tumour types remain refractive. Oncolytic viruses, such as those based on adenovirus, have great potential as anti-cancer agents and have seen a resurgence of interest in recent years. Amongst their many advantages is their ability to induce immunogenic cell death (ICD) of infected tumour cells, thus providing the alluring potential to synergise with immunotherapies by turning immunologically "cold" tumours "hot". Additionally, enhanced immune mediated cell killing can be promoted through the local overexpression of immunological transgenes, encoded from within the engineered viral genome. To achieve this full potential requires the development of refined, tumour selective "precision virotherapies" that are extensively engineered to prevent off-target up take via native routes of infection and targeted to infect and replicate uniquely within malignantly transformed cells. Here, we review the latest advances towards this holy grail within the adenoviral field.
Collapse
Affiliation(s)
| | | | - Alan L. Parker
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (T.G.C.); (E.A.B.)
| |
Collapse
|
96
|
Li X, Hu K, Liu W, Wei Y, Sha R, Long Y, Han Y, Sun P, Wu H, Li G, Tang G, Huang S. Synthesis and evaluation of [ 18F]FP-Lys-GE11 as a new radiolabeled peptide probe for epidermal growth factor receptor (EGFR) imaging. Nucl Med Biol 2020; 90-91:84-92. [PMID: 33189948 DOI: 10.1016/j.nucmedbio.2020.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/27/2020] [Accepted: 10/21/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The epidermal growth factor receptor (EGFR) has emerged as an attractive target in the treatment of various cancers. Radiolabeled small molecules, antibodies, and peptides that specifically target EGFR are promising probes for tumor imaging to guide personalized treatment with EGFR-targeted drugs. This study aimed to radiolabel GE11 (an EGFR-specific targeting peptide) with 18-fluorine to develop a new EGFR-targeting positron emission tomography (PET) probe, [18F]FP-Lys-GE11, for imaging tumors overexpressing EGFR. METHODS [18F]FP-Lys-GE11 was produced by radiolabeling a GE11 peptide with the prosthetic group 4-nitrophenyl-2-[18F]fluoropropionate ([18F]NFP). Stability in PBS and mice serum, affinity for A431 cell line, U87 and PC-3 cells uptake and blocking studies, and biodistribution of [18F]FP-Lys-GE11 were determined. 2 h dynamic and static PET scans of probe for tumor-bearing mice normal and inhibition uptake were performed. RESULTS [18F]FP-Lys-GE11 was stable in PBS and mice serum. The Kd and Bmax values of probe for A431 were 42.43 ± 3.75 nM and 3383 ± 81.73 CPM, respectively. In cell uptake and blocking experiments, a significant reduction in radioactivity accumulation (over 4-fold) was observed by blocking U87 and PC-3 cells with unlabeled peptide. PET imaging of U87 and PC-3 tumor-bearing mice revealed clear tumor imaging (tumor radioactivity accumulation was 3.48 ± 0.44 and 3.68 ± 0.76%ID/g respectively, tumor-to-muscle ratio was 3.45 ± 0.43 and 3.64 ± 0.76 respectively). Blocking imaging revealed that the U87 tumor uptake was significantly inhibited (2.21 ± 0.41%ID/g). The biodistribution and dynamic PET imaging showed that [18F]FP-Lys-GE11 was mainly excreted by the kidneys and the rest was excreted through the bile and intestines. CONCLUSION The current results showed that [18F]FP-Lys-GE11was a good radiolabeled peptide probe for EGFR overexpression tumor's imaging.
Collapse
Affiliation(s)
- Xueli Li
- Medical Imaging Profession, Nanfang Hospital, Southern Medical University, Guangdong 510515, China
| | - Kongzhen Hu
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangdong 510515, China
| | - Wenfeng Liu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Yuanfeng Wei
- Medical Imaging Profession, Nanfang Hospital, Southern Medical University, Guangdong 510515, China
| | - Runhua Sha
- Medical Imaging Profession, Nanfang Hospital, Southern Medical University, Guangdong 510515, China
| | - Yongxuan Long
- Medical Imaging Profession, Nanfang Hospital, Southern Medical University, Guangdong 510515, China
| | - Yanjiang Han
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangdong 510515, China
| | - Penhui Sun
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangdong 510515, China
| | - Hubing Wu
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangdong 510515, China
| | - Guiping Li
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangdong 510515, China
| | - Ganghua Tang
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangdong 510515, China
| | - Shun Huang
- Medical Imaging Profession, Nanfang Hospital, Southern Medical University, Guangdong 510515, China; Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangdong 510515, China.
| |
Collapse
|
97
|
Hartzell EJ, Lieser RM, Sullivan MO, Chen W. Modular Hepatitis B Virus-like Particle Platform for Biosensing and Drug Delivery. ACS NANO 2020; 14:12642-12651. [PMID: 32924431 DOI: 10.1021/acsnano.9b08756] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The hepatitis B virus-like particle (HBV VLP) is an attractive protein nanoparticle platform due to the availability of 240 modification sites for engineering purposes. Although direct protein insertion into the surface loop has been demonstrated, this decoration strategy is restricted by the size of the inserted protein moieties. Meanwhile, larger proteins can be decorated using chemical conjugations; yet these approaches perturb the integrity of more delicate proteins and can unfavorably orient the proteins, impairing active surface display. Herein, we aim to create a robust and highly modular method to produce smart HBV-based nanodevices by using the SpyCatcher/SpyTag system, which allows a wide range of peptides and proteins to be conjugated directly and simply onto the modified HBV capsids in a controlled and biocompatible manner. Our technology allows the modular surface modification of HBV VLPs with multiple components, which provides signal amplification, increased targeting avidity, and high therapeutic payload incorporation. We have achieved a yield of over 200 mg/L for these engineered HBV VLPs and demonstrated the flexibility of this platform in both biosensing and drug delivery applications. The ability to decorate over 200 nanoluciferases per VLP improved detection signal by over 1500-fold, such that low nanomolar levels of thrombin could be detected by the naked eye. Meanwhile, a dimeric prodrug-activating enzyme was loaded without cross-linking particles by coexpressing orthogonally labeled monomers. This along with a epidermal growth factor receptor-binding peptide enabled tunable uptake of HBV VLPs into inflammatory breast cancer cells, leading to efficient suicide enzyme delivery and cell killing.
Collapse
Affiliation(s)
- Emily J Hartzell
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Rachel M Lieser
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| |
Collapse
|
98
|
Chu W, Prodromou R, Day KN, Schneible JD, Bacon KB, Bowen JD, Kilgore RE, Catella CM, Moore BD, Mabe MD, Alashoor K, Xu Y, Xiao Y, Menegatti S. Peptides and pseudopeptide ligands: a powerful toolbox for the affinity purification of current and next-generation biotherapeutics. J Chromatogr A 2020; 1635:461632. [PMID: 33333349 DOI: 10.1016/j.chroma.2020.461632] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023]
Abstract
Following the consolidation of therapeutic proteins in the fight against cancer, autoimmune, and neurodegenerative diseases, recent advancements in biochemistry and biotechnology have introduced a host of next-generation biotherapeutics, such as CRISPR-Cas nucleases, stem and car-T cells, and viral vectors for gene therapy. With these drugs entering the clinical pipeline, a new challenge lies ahead: how to manufacture large quantities of high-purity biotherapeutics that meet the growing demand by clinics and biotech companies worldwide. The protein ligands employed by the industry are inadequate to confront this challenge: while featuring high binding affinity and selectivity, these ligands require laborious engineering and expensive manufacturing, are prone to biochemical degradation, and pose safety concerns related to their bacterial origin. Peptides and pseudopeptides make excellent candidates to form a new cohort of ligands for the purification of next-generation biotherapeutics. Peptide-based ligands feature excellent target biorecognition, low or no toxicity and immunogenicity, and can be manufactured affordably at large scale. This work presents a comprehensive and systematic review of the literature on peptide-based ligands and their use in the affinity purification of established and upcoming biological drugs. A comparative analysis is first presented on peptide engineering principles, the development of ligands targeting different biomolecular targets, and the promises and challenges connected to the industrial implementation of peptide ligands. The reviewed literature is organized in (i) conventional (α-)peptides targeting antibodies and other therapeutic proteins, gene therapy products, and therapeutic cells; (ii) cyclic peptides and pseudo-peptides for protein purification and capture of viral and bacterial pathogens; and (iii) the forefront of peptide mimetics, such as β-/γ-peptides, peptoids, foldamers, and stimuli-responsive peptides for advanced processing of biologics.
Collapse
Affiliation(s)
- Wenning Chu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Raphael Prodromou
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kevin N Day
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - John D Schneible
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kaitlyn B Bacon
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - John D Bowen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Ryan E Kilgore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Carly M Catella
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Brandyn D Moore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Matthew D Mabe
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kawthar Alashoor
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, NY 14642
| | - Yiman Xu
- College of Material Science and Engineering, Donghua University, 201620 Shanghai, People's Republic of China
| | - Yuanxin Xiao
- College of Textile, Donghua University, Songjiang District, Shanghai, 201620, People's Republic of China
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606.
| |
Collapse
|
99
|
Massaro C, Sgueglia G, Frattolillo V, Baglio SR, Altucci L, Dell’Aversana C. Extracellular Vesicle-Based Nucleic Acid Delivery: Current Advances and Future Perspectives in Cancer Therapeutic Strategies. Pharmaceutics 2020; 12:pharmaceutics12100980. [PMID: 33081417 PMCID: PMC7589909 DOI: 10.3390/pharmaceutics12100980] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs) are sophisticated and sensitive messengers released by cells to communicate with and influence distant and neighboring cells via selective transfer of bioactive content, including protein lipids and nucleic acids. EVs have therefore attracted broad interest as new and refined potential therapeutic systems in many diseases, including cancer, due to their low immunogenicity, non-toxicity, and elevated bioavailability. They might serve as safe and effective vehicles for the transport of therapeutic molecules to specific tissues and cells. In this review, we focus on EVs as a vehicle for gene therapy in cancer. We describe recent developments in EV engineering to achieve efficient intracellular delivery of cancer therapeutics and avoid off-target effects, to provide an overview of the potential applications of EV-mediated gene therapy and the most promising biomedical advances.
Collapse
Affiliation(s)
- Crescenzo Massaro
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy; (C.M.); (G.S.); (V.F.)
| | - Giulia Sgueglia
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy; (C.M.); (G.S.); (V.F.)
| | - Victoria Frattolillo
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy; (C.M.); (G.S.); (V.F.)
| | - S. Rubina Baglio
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, 1081HV Amsterdam, The Netherlands;
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy; (C.M.); (G.S.); (V.F.)
- Correspondence: (L.A.); (C.D.); Tel.: +39-081-5667569 (L.A.); +39-081-5667564 (C.D.)
| | - Carmela Dell’Aversana
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy; (C.M.); (G.S.); (V.F.)
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS)-National Research Council (CNR), Via Sergio Pansini 5, 80131 Naples, Italy
- Correspondence: (L.A.); (C.D.); Tel.: +39-081-5667569 (L.A.); +39-081-5667564 (C.D.)
| |
Collapse
|
100
|
Lieser RM, Yur D, Sullivan MO, Chen W. Site-Specific Bioconjugation Approaches for Enhanced Delivery of Protein Therapeutics and Protein Drug Carriers. Bioconjug Chem 2020; 31:2272-2282. [DOI: 10.1021/acs.bioconjchem.0c00456] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Rachel M. Lieser
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Daniel Yur
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Millicent O. Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| |
Collapse
|