51
|
Ye Q, Wang Y, Shen S, Xu C, Wang J. Biomaterials-Based Delivery of Therapeutic Antibodies for Cancer Therapy. Adv Healthc Mater 2021; 10:e2002139. [PMID: 33870637 DOI: 10.1002/adhm.202002139] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/05/2021] [Indexed: 12/19/2022]
Abstract
Considerable breakthroughs in the treatment of malignant tumors using antibody drugs, especially immunomodulating monoclonal antibodies (mAbs), have been made in the past decade. Despite technological advancements in antibody design and manufacture, multiple challenges face antibody-mediated cancer therapy, such as instability in vivo, poor tumor penetration, limited response rate, and undesirable off-target cytotoxicity. In recent years, an increasing number of biomaterials-based delivery systems have been reported to enhance the antitumor efficacy of antibody drugs. This review summarizes the advances and breakthroughs in integrating biomaterials with therapeutic antibodies for enhanced cancer therapy. A brief introduction to the principal mechanism of antibody-based cancer therapy is first established, and then various antibody immobilization strategies are provided. Finally, the current state-of-the-art in biomaterials-based antibody delivery systems and their applications in cancer treatment are summarized, highlighting how the delivery systems augment the therapeutic efficacy of antibody drugs. The outlook and perspective on biomaterials-based delivery of antitumor antibodies are also discussed.
Collapse
Affiliation(s)
- Qian‐Ni Ye
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
| | - Yue Wang
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Shenzhen Bay Laboratory Shenzhen 518132 P. R. China
| | - Song Shen
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education South China University of Technology Guangzhou 510006 P. R. China
| | - Cong‐Fei Xu
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education South China University of Technology Guangzhou 510006 P. R. China
| | - Jun Wang
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction South China University of Technology Guangzhou 510006 P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction South China University of Technology Guangzhou 510006 P. R. China
| |
Collapse
|
52
|
Hara D, Tao W, Totiger TM, Pourmand A, Dogan N, Ford JC, Shi J, Pollack A. Prostate Cancer Targeted X-Ray Fluorescence Imaging via Gold Nanoparticles Functionalized With Prostate-Specific Membrane Antigen (PSMA). Int J Radiat Oncol Biol Phys 2021; 111:220-232. [PMID: 33964351 DOI: 10.1016/j.ijrobp.2021.04.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/06/2021] [Accepted: 04/25/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE The gold nanoparticle (GNP) as a promising theranostic probe has been increasingly studied. The tumor-targeting efficiency of GNPs is crucial to increase the therapeutic ratio. In this study, we developed PSMA-targeted GNPs to enhance GNP uptake in prostate cancer and developed an x-ray fluorescence imaging system to noninvasively monitor and assess GNP delivery. METHODS AND MATERIALS For targeted therapy of prostate cancer, anti-prostate-specific membrane antigen (PSMA) antibodies were conjugated onto PEGylated GNPs through 1-ethyl-3-(-3-dimethylaminopropyl) carbodiimide (EDC) and N-hydroxysuccinimide (NHS) (EDC/NHS) chemistry. In vivo imaging was implemented using an in-house-developed dual-modality computed tomography (CT) and x-ray fluorescence CT (XFCT) system on mice bearing subcutaneous LNCaP prostate tumors. After intravenous administration of GNPs (15 mg/mL, 200 μL), the x-ray fluorescence signals from the tumor were collected at various time points (5 minutes to approximately 30 hours) for GNP pharmacokinetics analysis. At 24 hours after administration, x-ray fluorescence projection (XRFproj) and XFCT imaging were conducted to evaluate the prostate tumor uptake of active- and passive-targeting GNPs. Inductively coupled plasma mass spectrometry analysis was adopted as a benchmark to verify the quantification accuracy of XRFproj/XFCT imaging. RESULTS Fluorescence microscopic imaging confirmed the enhanced (approximately 4 times) targeting efficiency of PSMA-targeted GNPs in vitro. The pharmacokinetics analysis showed enhanced tumor uptake/retention of PSMA-targeted GNPs and revealed that the peak tumor accumulation appeared at approximately 24 hours after intravenous administration. Both XRFproj and XFCT imaging presented their accuracy in quantifying GNPs within tumors noninvasively. Moreover, XFCT imaging verified its unique capabilities to simultaneously determine the heterogeneous spatial distribution and the concentration of GNPs within tumors in vivo. CONCLUSIONS In conjunction with PSMA-targeted GNPs, XRFproj/XFCT would be a highly sensitive tool for targeted imaging of prostate cancer, benefiting the elucidation of mechanisms of GNP-assisted prostate-cancer therapy.
Collapse
Affiliation(s)
- Daiki Hara
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, Florida; Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, Florida
| | - Wensi Tao
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Tulasigeri M Totiger
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Ali Pourmand
- Rosenstiel School of Marine and Atmospheric Sciences, University of Miami, Miami, Florida
| | - Nesrin Dogan
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, Florida
| | - John Chetley Ford
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, Florida; Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, Florida.
| | - Junwei Shi
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, Florida.
| | - Alan Pollack
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
53
|
Peng Y, Rabin C, Walgama CT, Pollok NE, Smith L, Richards I, Crooks RM. Silver Nanocubes as Electrochemical Labels for Bioassays. ACS Sens 2021; 6:1111-1119. [PMID: 33439628 DOI: 10.1021/acssensors.0c02377] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Here, we report on the use of 40 ± 4 nm silver nanocubes (AgNCs) as electrochemical labels in bioassays. The model metalloimmunoassay combines galvanic exchange (GE) and anodic stripping voltammetry (ASV). The results show that a lower limit of detection is achieved by simply changing the shape of the Ag label yielding improved GE with AgNCs when compared to GE with spherical silver nanoparticles (sAgNPs). Specifically, during GE between electrogenerated Au3+ and the Ag labels, a thin shell of Au forms on the surface of the NP. This shell is more porous when GE proceeds on AgNCs compared to sAgNPs, and therefore, more exchange occurs when using AgNCs. ASV results show that the Ag collection efficiency (AgCE%) is increased by up to ∼57% when using AgNCs. When the electrochemical system is fully optimized, the limit of detection is 0.1 pM AgNCs, which is an order of magnitude lower than that of sAgNP labels.
Collapse
Affiliation(s)
- Yi Peng
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Charlie Rabin
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Charuksha T. Walgama
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Nicole E. Pollok
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Leilani Smith
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Ian Richards
- Interactives Executive Excellence LLC, Austin, Texas 78733 United States
| | - Richard M. Crooks
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| |
Collapse
|
54
|
António M, Vitorino R, Daniel-da-Silva AL. Gold nanoparticles-based assays for biodetection in urine. Talanta 2021; 230:122345. [PMID: 33934794 DOI: 10.1016/j.talanta.2021.122345] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/24/2022]
Abstract
Urine is a biofluid easy to collect through a non-invasive technique that allows collecting a large volume of sample. The use of urine for disease diagnosis is not yet well explored. However, it has gained attention over the last three years. It has been applied in the diagnosis of several illnesses such as kidney disease, bladder cancer, prostate cancer and cardiovascular diseases. In the last decade, gold nanoparticles (Au NPs) have attracted attention in biosensors' development for the diagnosis of diseases due to their electrical and optical properties, ability to conjugate with biomolecules, high sensitivity, and selectivity. Therefore, this article aims to present a comprehensive view of state of the art on the advances made in the quantification of analytes in urinary samples using AuNPs based assays, with a focus on protein analysis. The type of diagnosis methods, the Au NPs synthesis approaches and the strategies for surface modification aiming at selectivity towards the different targets are highlighted.
Collapse
Affiliation(s)
- Maria António
- CICECO-Aveiro Institute of Materials, Chemistry Department, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rui Vitorino
- iBiMED-Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, 3810-193, Portugal; Department of Surgery and Physiology, Cardiovascular R&D Center, Faculty of Medicine of the University of Porto, Alameda Professor Hernâni Monteiro, 4200-319, Porto, Portugal; LAQV-REQUIMTE, Chemistry Department, University of Aveiro, Aveiro, Portugal.
| | - Ana L Daniel-da-Silva
- CICECO-Aveiro Institute of Materials, Chemistry Department, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
55
|
Lin X, O'Reilly Beringhs A, Lu X. Applications of Nanoparticle-Antibody Conjugates in Immunoassays and Tumor Imaging. AAPS J 2021; 23:43. [PMID: 33718979 PMCID: PMC7956929 DOI: 10.1208/s12248-021-00561-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Modern diagnostic technologies rely on both in vitro and in vivo modalities to provide a complete understanding of the clinical state of a patient. Nanoparticle-antibody conjugates have emerged as promising systems to confer increased sensitivity and accuracy for in vitro diagnostics (e.g., immunoassays). Meanwhile, in vivo applications have benefited from the targeting ability of nanoparticle-antibody conjugates, as well as payload flexibility and tailored biodistribution. This review provides an encompassing overview of nanoparticle-antibody conjugates, from chemistry to applications in medical immunoassays and tumor imaging, highlighting the underlying principles and unique features of relevant preclinical applications employing commonly used imaging modalities (e.g., optical/photoacoustics, positron-emission tomography, magnetic resonance imaging, X-ray computed tomography).
Collapse
Affiliation(s)
- Xinhao Lin
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut, USA
| | | | - Xiuling Lu
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut, USA.
| |
Collapse
|
56
|
Jiang CT, Chen KG, Liu A, Huang H, Fan YN, Zhao DK, Ye QN, Zhang HB, Xu CF, Shen S, Xiong MH, Du JZ, Yang XZ, Wang J. Immunomodulating nano-adaptors potentiate antibody-based cancer immunotherapy. Nat Commun 2021; 12:1359. [PMID: 33649336 PMCID: PMC7921676 DOI: 10.1038/s41467-021-21497-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Modulating effector immune cells via monoclonal antibodies (mAbs) and facilitating the co-engagement of T cells and tumor cells via chimeric antigen receptor- T cells or bispecific T cell-engaging antibodies are two typical cancer immunotherapy approaches. We speculated that immobilizing two types of mAbs against effector cells and tumor cells on a single nanoparticle could integrate the functions of these two approaches, as the engineered formulation (immunomodulating nano-adaptor, imNA) could potentially associate with both cells and bridge them together like an 'adaptor' while maintaining the immunomodulatory properties of the parental mAbs. However, existing mAbs-immobilization strategies mainly rely on a chemical reaction, a process that is rough and difficult to control. Here, we build up a versatile antibody immobilization platform by conjugating anti-IgG (Fc specific) antibody (αFc) onto the nanoparticle surface (αFc-NP), and confirm that αFc-NP could conveniently and efficiently immobilize two types of mAbs through Fc-specific noncovalent interactions to form imNAs. Finally, we validate the superiority of imNAs over the mixture of parental mAbs in T cell-, natural killer cell- and macrophage-mediated antitumor immune responses in multiple murine tumor models.
Collapse
Affiliation(s)
- Cheng-Tao Jiang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Kai-Ge Chen
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, PR China
| | - An Liu
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, PR China
| | - Hua Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Ya-Nan Fan
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Dong-Kun Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Qian-Ni Ye
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Hou-Bing Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Cong-Fei Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
- Shenzhen Bay Laboratory, Shenzhen, 518132, PR China
| | - Song Shen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China.
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China.
| | - Meng-Hua Xiong
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Jin-Zhi Du
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Xian-Zhu Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China.
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, PR China.
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
57
|
Hu J, Zhou S, Zeng L, Chen Q, Duan H, Chen X, Li X, Xiong Y. Hydrazide mediated oriented coupling of antibodies on quantum dot beads for enhancing detection performance of immunochromatographic assay. Talanta 2021; 223:121723. [DOI: 10.1016/j.talanta.2020.121723] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/15/2020] [Accepted: 09/28/2020] [Indexed: 10/23/2022]
|
58
|
Nojoomi A, Jeon J, Yum K. 2D material programming for 3D shaping. Nat Commun 2021; 12:603. [PMID: 33504805 PMCID: PMC7841157 DOI: 10.1038/s41467-021-20934-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 01/04/2021] [Indexed: 12/02/2022] Open
Abstract
Two-dimensional (2D) growth-induced 3D shaping enables shape-morphing materials for diverse applications. However, quantitative design of 2D growth for arbitrary 3D shapes remains challenging. Here we show a 2D material programming approach for 3D shaping, which prints hydrogel sheets encoded with spatially controlled in-plane growth (contraction) and transforms them to programmed 3D structures. We design 2D growth for target 3D shapes via conformal flattening. We introduce the concept of cone singularities to increase the accessible space of 3D shapes. For active shape selection, we encode shape-guiding modules in growth that direct shape morphing toward target shapes among isometric configurations. Our flexible 2D printing process enables the formation of multimaterial 3D structures. We demonstrate the ability to create 3D structures with a variety of morphologies, including automobiles, batoid fish, and real human face. Two-dimensional (2D) growth-induced 3D shaping enables shape-morphing materials for diverse applications but quantitative design of 2D growth for arbitrary 3D shapes remains challenging. Here, the authors show a 2D material programming approach for arbitrary 3D shaping, which prints hydrogel sheets encoded with spatially controlled in-plane growth and transforms them to programmed 3D structures.
Collapse
Affiliation(s)
- Amirali Nojoomi
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, TX, USA
| | - Junha Jeon
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, TX, USA
| | - Kyungsuk Yum
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, TX, USA.
| |
Collapse
|
59
|
Nanomaterials for Protein Delivery in Anticancer Applications. Pharmaceutics 2021; 13:pharmaceutics13020155. [PMID: 33503889 PMCID: PMC7910976 DOI: 10.3390/pharmaceutics13020155] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 12/16/2022] Open
Abstract
Nanotechnology platforms, such as nanoparticles, liposomes, dendrimers, and micelles have been studied extensively for various drug deliveries, to treat or prevent diseases by modulating physiological or pathological processes. The delivery drug molecules range from traditional small molecules to recently developed biologics, such as proteins, peptides, and nucleic acids. Among them, proteins have shown a series of advantages and potential in various therapeutic applications, such as introducing therapeutic proteins due to genetic defects, or used as nanocarriers for anticancer agents to decelerate tumor growth or control metastasis. This review discusses the existing nanoparticle delivery systems, introducing design strategies, advantages of using each system, and possible limitations. Moreover, we will examine the intracellular delivery of different protein therapeutics, such as antibodies, antigens, and gene editing proteins into the host cells to achieve anticancer effects and cancer vaccines. Finally, we explore the current applications of protein delivery in anticancer treatments.
Collapse
|
60
|
Huang T, Li S, Fang J, Li F, Tu S. Antibody-activated trans-endothelial delivery of mesoporous organosilica nanomedicine augments tumor extravasation and anti-cancer immunotherapy. Bioact Mater 2021; 6:2158-2172. [PMID: 33511314 PMCID: PMC7815474 DOI: 10.1016/j.bioactmat.2020.12.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 11/18/2022] Open
Abstract
Tumor vasculature constitutes a formidable hurdle for the efficient delivery of cancer nanomedicine into tumors. The leverage of passive pathway through inter-endothelial gaps in tumor blood vessels might account for limited extravasation of nanomedicine into tumor microenvironment (TME). Herein, Annexin A1 antibody-installed mesoporous organosilica nanoplatforms carrying immunotherapeutics of anti-PD-L1 antibody (aPD-L1) and Indoximod are developed to target at caveolar Annexin-A1 protein of luminal endothelial cells and to trigger the active trans-endothelial transcytosis of nanomedicine mediated by caveolae. Such strategy enables rapid nanomedicine extravasation across tumor endothelium and relatively extensive accumulation in tumor interstitium. aPD-L1 and Indoximod release from aPD/IND@MON-aANN in a reduction-responsive manner and synergistically facilitate the intratumoral infiltration of cytotoxic T lymphocytes and reverse the immunosuppressive TME, thus demonstrating substantial anti-tumor efficacy in subcutaneous 4T1 breast tumors and remarkable anti-metastatic capacity to extend the survival of 4T1 tumor metastasis model. Moreover, aPD/IND@MON-aANN nanomedicine also exhibits distinct superiority over the combination therapy of free drugs to potently attenuate the progression of urethane-induced orthotopic lung cancers. Collectively, aPD/IND@MON-aANN nanoplatforms with boosted delivery efficiency via antibody-activated trans-endothelial pathway and enhanced immunotherapeutic efficacy provides perspectives for the development of cancer nanomedicines. The nanomedicine overcomes tumor vascular barrier by active transcytosis via caveolae initiated by the conjugated aANXA1. The nanoplatform responsively releases aPD-L1 and Indoximod to synergistically improve the efficacy of immunotherapy. The nanomedicine shows anti-tumor capacity in mice breast cancers and lung cancers.
Collapse
Affiliation(s)
- Tinglei Huang
- Department of Oncology, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shuang Li
- Department of Stomatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jianchen Fang
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Fuli Li
- Department of Oncology, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shuiping Tu
- Department of Oncology, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Corresponding author.
| |
Collapse
|
61
|
Katoozi D, Clayton AHA, Moss DJ, Chon JWM. Uptake quantification of gold nanoparticles inside of cancer cells using high order image correlation spectroscopy. BIOMEDICAL OPTICS EXPRESS 2021; 12:539-552. [PMID: 33659088 PMCID: PMC7899503 DOI: 10.1364/boe.417321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 06/12/2023]
Abstract
The application of gold nanoparticles (AuNPs) in cancer therapeutics and diagnostics has recently reached a clinical level. Functional use of the AuNP in theranostics first requires effective uptake into the cells, but accurate quantification of AuNPs cellular uptake in real-time is still a challenge due to the destructive nature of existing characterization methods. The optical imaging-based quantification method is highly desirable. Here, we propose the use of high-order image correlation spectroscopy (HICS) as an optical imaging-based nanoparticle quantification technique. Coupled with dark field microscopy (DFM), a non-destructive and easy quantification method could be achieved. We demonstrate HICS analysis on 80 nm AuNPs coated with cetyltrimethylammonium bromide (CTAB) uptake in HeLa cells to calculate the percentage of aggregate species (dimer) in the total uptake and their relative scattering quantum yield inside the cells, the details of which are not available with other quantification techniques. The total particle uptake kinetics measured were in a reasonable agreement with the literature.
Collapse
|
62
|
Wei Z, Xi Z, Vlasov S, Ayala J, Xia X. Nanocrystals of platinum-group metals as peroxidase mimics for in vitro diagnostics. Chem Commun (Camb) 2020; 56:14962-14975. [PMID: 33188672 DOI: 10.1039/d0cc06575g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Peroxidase mimics of nanoscale materials as alternatives to natural peroxidases have found widespread uses in biomedicine. Among various types of peroxidase mimics, platinum-group metal (PGM) nanocrystals have drawn considerable attention in recent years due to their superior properties. Particularly, PGM nanocrystals display high catalytic efficiencies, allow for facile surface modifications, and possess excellent stabilities. This feature article summarizes our recent work on development of PGM nanocrystals as peroxidase mimics and exploration of their applications in in vitro diagnostics. We begin with a brief introduction to controlled synthesis of PGM nanocrystals in solution phase. We then elaborate on a variety of physicochemical parameters that can be carefully tuned to optimize the peroxidase-like properties of PGM nanocrystals. Then, we highlight the applications of PGM nanocrystals in different in vitro diagnostic platforms. We conclude this article with personal perspectives on future research directions in this emerging field, where challenges and opportunities are remarked.
Collapse
Affiliation(s)
- Zhiyuan Wei
- Department of Chemistry, University of Central Florida, Orlando, Florida 32816, USA.
| | | | | | | | | |
Collapse
|
63
|
Kukkar D, Kukkar P, Kumar V, Hong J, Kim KH, Deep A. Recent advances in nanoscale materials for antibody-based cancer theranostics. Biosens Bioelectron 2020; 173:112787. [PMID: 33190049 DOI: 10.1016/j.bios.2020.112787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/08/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
The quest for advanced management tools or options of various cancers has been on the rise to efficiently reduce their risks of mortality without the demerits of conventional treatments (e.g., undesirable side effects of the medications on non-target tissues, non-targeted distribution, slow clearance of the administered drugs, and the development of drug resistance over the duration of therapy). In this context, nanomaterials-antibody conjugates can offer numerous advantages in the development of cancer theranostics over conventional delivery systems (e.g., highly specific and enhanced biodistribution of the drug in targeted tissues, prolonged systemic circulation, low toxicity, and minimally invasive molecular imaging). This review comprehensively discusses and evaluates recent advances in the application of nanomaterial-antibody bioconjugates for cancer theranostics for the further advancement in the control of diverse cancerous diseases. Further, discussion is expanded to cover the various challenges and limitations associated with the design and development of nanomaterial-antibody conjugates applicable towards better management of cancer.
Collapse
Affiliation(s)
- Deepak Kukkar
- Department of Nanotechnology, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, 140406, India
| | - Preeti Kukkar
- Department of Chemistry, Mata Gujri College, Fatehgarh Sahib, Punjab, 140406, India
| | - Vanish Kumar
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Punjab, 140306, India
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, Seoul, 04763 Republic of Korea.
| | - Akash Deep
- Central Scientific Instruments Organization (CSIR-CSIO), Sector 30 C, Chandigarh, 160030, India.
| |
Collapse
|
64
|
Hyperbranched Polyglycerols as Robust Up-Conversion Nanoparticle Coating Layer for Feasible Cell Imaging. Polymers (Basel) 2020; 12:polym12112592. [PMID: 33158226 PMCID: PMC7694285 DOI: 10.3390/polym12112592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 01/18/2023] Open
Abstract
Owing to the wide spectrum of excitation wavelengths of up-conversion nanoparticles (UCNPs) by precisely regulating the percentage of doping elements, UCNPs have been emerging as bioimaging agents. The key drawback of UCNPs is their poor dispersibility in aqueous solution and it is hard to introduce the chemical versatility of function groups. In our study, we present a robust and feasible UCNP modification approach by introducing hyperbranched polyglycerols (hbPGs) as a coating layer. When grafted by hbPGs, the solubility and biocompatibility of UCNPs are significantly improved. Moreover, we also systematically investigated and optimized the chemical modification approach of amino acids or green fluorescence protein (GFP), respectively, grafting onto hbPGs and hbPGs-g-UCNP by oxidizing the vicinal diol to be an aldehyde group, which reacts more feasibly with amino-containing functional molecules. Then, we investigated the drug-encapsulating properties of hbPGs-Arg with DOX and cell imaging of GFP-grafted hbPGs-g-UCNP, respectively. The excellent cell imaging in tumor cells indicated that hbPG-modification of UCNPs displayed potential for applications in drug delivery and disease diagnosis.
Collapse
|
65
|
Adak AK, Huang KT, Li PJ, Fan CY, Lin PC, Hwang KC, Lin CC. Regioselective S N2-Type Reaction for the Oriented and Irreversible Immobilization of Antibodies to a Glass Surface Assisted by Boronate Formation. ACS APPLIED BIO MATERIALS 2020; 3:6756-6767. [PMID: 35019340 DOI: 10.1021/acsabm.0c00700] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Antibodies have exquisite specificities for molecular recognition, which have led to their incorporation into array sensors that are crucial for research, diagnostic, and therapeutic applications. Many of these platforms rely heavily on surface-bound reactive groups to covalently tether antibodies to solid substrates; however, this strategy is hindered by a lack of orientation control over antibody immobilization. Here, we report a mild electrophilic phenylsulfonate (tosylate) ester-containing boronic acid affinity ligand for attaching antibodies to glass slides. A high level of antibody coupling located near the Fc region of the boronated antibody complex could be achieved by the proximal nucleophilic amino acid driven substitution reaction at the phenylsulfonate center. This enabled the full-length antibodies to be permanently tethered onto surfaces in an oriented manner. The advantages of this strategy were demonstrated through the individual and multiplex detection of protein and serum biomarkers. This strategy not only confers stability to the immobilized antibodies but also presents a different direction for the irreversible attachment of antibodies to solid supports in an orientation-controlled way.
Collapse
Affiliation(s)
- Avijit K Adak
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Kuan-Ting Huang
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Pei-Jhen Li
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chen-Yo Fan
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Po-Chiao Lin
- Department of Chemistry, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Kuo-Chu Hwang
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chun-Cheng Lin
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.,Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
66
|
Yu C, Schimelman J, Wang P, Miller KL, Ma X, You S, Guan J, Sun B, Zhu W, Chen S. Photopolymerizable Biomaterials and Light-Based 3D Printing Strategies for Biomedical Applications. Chem Rev 2020; 120:10695-10743. [PMID: 32323975 PMCID: PMC7572843 DOI: 10.1021/acs.chemrev.9b00810] [Citation(s) in RCA: 248] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Since the advent of additive manufacturing, known commonly as 3D printing, this technology has revolutionized the biofabrication landscape and driven numerous pivotal advancements in tissue engineering and regenerative medicine. Many 3D printing methods were developed in short course after Charles Hull first introduced the power of stereolithography to the world. However, materials development was not met with the same enthusiasm and remained the bottleneck in the field for some time. Only in the past decade has there been deliberate development to expand the materials toolbox for 3D printing applications to meet the true potential of 3D printing technologies. Herein, we review the development of biomaterials suited for light-based 3D printing modalities with an emphasis on bioprinting applications. We discuss the chemical mechanisms that govern photopolymerization and highlight the application of natural, synthetic, and composite biomaterials as 3D printed hydrogels. Because the quality of a 3D printed construct is highly dependent on both the material properties and processing technique, we included a final section on the theoretical and practical aspects behind light-based 3D printing as well as ways to employ that knowledge to troubleshoot and standardize the optimization of printing parameters.
Collapse
Affiliation(s)
- Claire Yu
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jacob Schimelman
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Pengrui Wang
- Materials Science and Engineering Program, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Kathleen L Miller
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Xuanyi Ma
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Shangting You
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jiaao Guan
- Department of Electrical and Computer Engineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Bingjie Sun
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Wei Zhu
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Shaochen Chen
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Materials Science and Engineering Program, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Chemical Engineering Program, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
67
|
Zhang L, Mazouzi Y, Salmain M, Liedberg B, Boujday S. Antibody-Gold Nanoparticle Bioconjugates for Biosensors: Synthesis, Characterization and Selected Applications. Biosens Bioelectron 2020; 165:112370. [DOI: 10.1016/j.bios.2020.112370] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 01/22/2023]
|
68
|
Li Y, Li P, Li R, Xu Q. Intracellular Antibody Delivery Mediated by Lipids, Polymers, and Inorganic Nanomaterials for Therapeutic Applications. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000178] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| | - Peixuan Li
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| | - Raissa Li
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| | - Qiaobing Xu
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| |
Collapse
|
69
|
Nanoparticles as labels of specific-recognition reactions for the determination of biomolecules by inductively coupled plasma-mass spectrometry. Anal Chim Acta 2020; 1128:251-268. [DOI: 10.1016/j.aca.2020.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 02/08/2023]
|
70
|
Bounab Y, Eyer K, Dixneuf S, Rybczynska M, Chauvel C, Mistretta M, Tran T, Aymerich N, Chenon G, Llitjos JF, Venet F, Monneret G, Gillespie IA, Cortez P, Moucadel V, Pachot A, Troesch A, Leissner P, Textoris J, Bibette J, Guyard C, Baudry J, Griffiths AD, Védrine C. Dynamic single-cell phenotyping of immune cells using the microfluidic platform DropMap. Nat Protoc 2020; 15:2920-2955. [PMID: 32788719 DOI: 10.1038/s41596-020-0354-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 05/06/2020] [Indexed: 02/08/2023]
Abstract
Characterization of immune responses is currently hampered by the lack of systems enabling quantitative and dynamic phenotypic characterization of individual cells and, in particular, analysis of secreted proteins such as cytokines and antibodies. We recently developed a simple and robust microfluidic platform, DropMap, to measure simultaneously the kinetics of secretion and other cellular characteristics, including endocytosis activity, viability and expression of cell-surface markers, from tens of thousands of single immune cells. Single cells are compartmentalized in 50-pL droplets and analyzed using fluorescence microscopy combined with an immunoassay based on fluorescence relocation to paramagnetic nanoparticles aligned to form beadlines in a magnetic field. The protocol typically takes 8-10 h after preparation of microfluidic chips and chambers, which can be done in advance. By contrast, enzyme-linked immunospot (ELISPOT), flow cytometry, time-of-flight mass cytometry (CyTOF), and single-cell sequencing enable only end-point measurements and do not enable direct, quantitative measurement of secreted proteins. We illustrate how this system can be used to profile downregulation of tumor necrosis factor-α (TNF-α) secretion by single monocytes in septic shock patients, to study immune responses by measuring rates of cytokine secretion from single T cells, and to measure affinity of antibodies secreted by single B cells.
Collapse
Affiliation(s)
- Yacine Bounab
- BIOASTER Technology Research Institute, Lyon, France.,Laboratoire de Biochimie (LBC), École Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris Sciences et Lettres (PSL), CNRS UMR8231, Paris, France
| | - Klaus Eyer
- Laboratoire de Colloïdes et Matériaux Divisés (LCMD), École Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris Sciences et Lettres (PSL), CNRS UMR8231, Paris, France.,Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, Zurich, Switzerland
| | - Sophie Dixneuf
- Biological Microsystems and Advanced Optics Engineering Unit, BIOASTER Technology Research Institute, Paris, France
| | - Magda Rybczynska
- Laboratoire de Colloïdes et Matériaux Divisés (LCMD), École Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris Sciences et Lettres (PSL), CNRS UMR8231, Paris, France
| | - Cécile Chauvel
- Biological Microsystems and Advanced Optics Engineering Unit, BIOASTER Technology Research Institute, Paris, France
| | | | - Trang Tran
- Biological Microsystems and Advanced Optics Engineering Unit, BIOASTER Technology Research Institute, Paris, France
| | - Nathan Aymerich
- Laboratoire de Colloïdes et Matériaux Divisés (LCMD), École Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris Sciences et Lettres (PSL), CNRS UMR8231, Paris, France
| | - Guilhem Chenon
- Laboratoire de Colloïdes et Matériaux Divisés (LCMD), École Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris Sciences et Lettres (PSL), CNRS UMR8231, Paris, France
| | | | - Fabienne Venet
- EA7426-Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon-1 - HCL - bioMérieux, Lyon, France.,Immunology Laboratory, Hospices Civils de Lyon, Lyon, France
| | - Guillaume Monneret
- EA7426-Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon-1 - HCL - bioMérieux, Lyon, France.,Immunology Laboratory, Hospices Civils de Lyon, Lyon, France
| | - Iain A Gillespie
- Value, Evidence & Outcomes, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | | | - Virginie Moucadel
- EA7426-Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon-1 - HCL - bioMérieux, Lyon, France.,Medical Diagnostic Discovery Department (MD3), bioMérieux S.A., Lyon, France
| | - Alexandre Pachot
- Medical Diagnostic Discovery Department (MD3), bioMérieux S.A., Lyon, France
| | - Alain Troesch
- Biological Microsystems and Advanced Optics Engineering Unit, BIOASTER Technology Research Institute, Paris, France
| | - Philippe Leissner
- Biological Microsystems and Advanced Optics Engineering Unit, BIOASTER Technology Research Institute, Paris, France
| | - Julien Textoris
- EA7426-Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon-1 - HCL - bioMérieux, Lyon, France.,Medical Diagnostic Discovery Department (MD3), bioMérieux S.A., Lyon, France.,Anesthesiology and Critical Care Medicine, Hospices Civils de Lyon (HCL), Lyon, France
| | - Jérôme Bibette
- Laboratoire de Colloïdes et Matériaux Divisés (LCMD), École Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris Sciences et Lettres (PSL), CNRS UMR8231, Paris, France
| | - Cyril Guyard
- Biological Microsystems and Advanced Optics Engineering Unit, BIOASTER Technology Research Institute, Paris, France
| | - Jean Baudry
- Laboratoire de Colloïdes et Matériaux Divisés (LCMD), École Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris Sciences et Lettres (PSL), CNRS UMR8231, Paris, France.
| | - Andrew D Griffiths
- Laboratoire de Biochimie (LBC), École Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris Sciences et Lettres (PSL), CNRS UMR8231, Paris, France.
| | | |
Collapse
|
71
|
Nieto C, Vega MA, Martín del Valle EM. Trastuzumab: More than a Guide in HER2-Positive Cancer Nanomedicine. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1674. [PMID: 32859026 PMCID: PMC7557948 DOI: 10.3390/nano10091674] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023]
Abstract
HER2 overexpression, which occurs in a fifth of diagnosed breast cancers as well as in other types of solid tumors, has been traditionally linked to greater aggressiveness. Nevertheless, the clinical introduction of trastuzumab has helped to improve HER2-positive patients' outcomes. As a consequence, nanotechnology has taken advantage of the beneficial effects of the administration of this antibody and has employed it to develop HER2-targeting nanomedicines with promising therapeutic activity and limited toxicity. In this review, the molecular pathways that could be responsible for trastuzumab antitumor activity will be briefly summarized. In addition, since the conjugation strategies that are followed to develop targeting nanomedicines are essential to maintaining their efficacy and tolerability, the ones most employed to decorate drug-loaded nanoparticles and liposomes with trastuzumab will be discussed here. Thus, the advantages and disadvantages of performing this trastuzumab conjugation through adsorption or covalent bindings (through carbodiimide, maleimide, and click-chemistry) will be described, and several examples of targeting nanovehicles developed following these strategies will be commented on. Moreover, conjugation methods employed to synthesized trastuzumab-based antibody drug conjugates (ADCs), among which T-DM1 is well known, will be also examined. Finally, although trastuzumab-decorated nanoparticles and liposomes and trastuzumab-based ADCs have proven to have better selectivity and efficacy than loaded drugs, trastuzumab administration is sometimes related to side toxicities and the apparition of resistances. For this reason also, this review focuses at last on the important role that newer antibodies and peptides are acquiring these days in the development of HER2-targeting nanomedicines.
Collapse
Affiliation(s)
- Celia Nieto
- Departamento de Ingeniería Química, Facultad de Ciencias Químicas, Universidad de Salamanca, 37008 Salamanca, Spain;
| | | | - Eva M. Martín del Valle
- Departamento de Ingeniería Química, Facultad de Ciencias Químicas, Universidad de Salamanca, 37008 Salamanca, Spain;
| |
Collapse
|
72
|
Zhong W, Zhang X, Zhao M, Wu J, Lin D. Advancements in nanotechnology for the diagnosis and treatment of multiple myeloma. Biomater Sci 2020; 8:4692-4711. [PMID: 32779645 DOI: 10.1039/d0bm00772b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Multiple myeloma (MM), known as a tumor of plasma cells, is not only refractory but also has a high relapse rate, and is the second-most common hematologic tumor after lymphoma. It is often accompanied by multiple osteolytic damage, hypercalcemia, anemia, and renal insufficiency. In terms of diagnosis, conventional detection methods have many limitations, such as it is invasive and time-consuming and has low accuracy. Measures to change these limitations are urgently needed. At the therapeutic level, although the survival of MM continues to prolong with the advent of new drugs, MM remains incurable and has a high recurrence rate. With the development of nanotechnology, nanomedicine has become a powerful way to improve the current diagnosis and treatment of MM. In this review, the research progress and breakthroughs of nanomedicine in MM will be presented. Meanwhile, both superiorities and challenges of nanomedicine were discussed. As a new idea for the diagnosis and treatments of MM, nanomedicine will play a very important role in the research field of MM.
Collapse
Affiliation(s)
- Wenhao Zhong
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P.R. China.
| | | | | | | | | |
Collapse
|
73
|
Hristov DR, Pimentel AJ, Ujialele G, Hamad-Schifferli K. The Immunoprobe Aggregation State is Central to Dipstick Immunoassay Performance. ACS APPLIED MATERIALS & INTERFACES 2020; 12:34620-34629. [PMID: 32633115 DOI: 10.1021/acsami.0c08628] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
As new infectious disease outbreaks become more likely, it is important to be able to develop and deploy appropriate testing in time. Paper-based immunoassays are rapid, cheap, and easy to produce at scale and relatively user friendly but often suffer from low selectivity and sensitivity. Understanding the molecular mechanisms of paper immunoassays may help improve and hasten development and therefore production and market availability. Here, we study how the behavior of nanoparticle-antibody immunoprobes in paper dipstick immunoassays is impacted by synthesis strategy and surface chemistry architecture. We conjugate gold nanoparticles to polyclonal anti-immunoglobulin G (IgG) and anti-zika NS1 antibodies by electrostatic adsorption and N-hydroxysuccinimide (NHS) and hydrazide (Hz) chemistries. The immunoprobes were used in paper immunoassays and the effective affinity for the antigen was quantified from the test line intensities, as well as the distribution of the immunoprobes throughout the strips. The results show that nanoparticle colloidal stability, both post synthesis and during antigen binding, is a key factor and affects immunoassay results and performance, often through reduction or loss of signal.
Collapse
|
74
|
Fotoran WL, Kleiber N, Müntefering T, Liebau E, Wunderlich G. Production of glycosylphosphatidylinositol-anchored proteins for vaccines and directed binding of immunoliposomes to specific cell types. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20200032. [PMID: 32788917 PMCID: PMC7401668 DOI: 10.1590/1678-9199-jvatitd-2020-0032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/14/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Liposomes are highly useful carriers for delivering drugs or antigens. The association of glycosylphosphatidylinositol (GPI)-anchored proteins to liposomes potentially enhances the immunogenic effect of vaccine antigens by increasing their surface concentration. Furthermore, the introduction of a universal immunoglobulin-binding domain can make liposomes targetable to virtually any desired receptor for which antibodies exist. METHODS We developed a system for the production of recombinant proteins with GPI anchors and histidine tags and Strep-tags for simplified purification from cells. This system was applied to i) the green fluorescent protein (GFP) as a reporter, ii) the promising Plasmodium falciparum vaccine antigen PfRH5 and iii) a doubled immunoglobulin Fc-binding domain termed ZZ from protein A of Staphylococcus aureus. As the GPI-attachment domain, the C-terminus of murine CD14 was used. After the recovery of these three recombinant proteins from Chinese hamster ovary (CHO) cells and association with liposomes, their vaccine potential and ability to target the CD4 receptor on lymphocytes in ex vivo conditions were tested. RESULTS Upon immunization in mice, the PfRH5-GPI-loaded liposomes generated antibody titers of 103 to 104, and showed a 45% inhibitory effect on in vitro growth at an IgG concentration of 600 µg/mL in P. falciparum cultures. Using GPI-anchored ZZ to couple anti-CD4 antibodies to liposomes, we created immunoliposomes with a binding efficiency of 75% to CD4+ cells in splenocytes and minimal off-target binding. CONCLUSIONS Proteins are very effectively associated with liposomes via a GPI-anchor to form proteoliposome particles and these are useful for a variety of applications including vaccines and antibody-mediated targeting of liposomes. Importantly, the CHO-cell and GPI-tagged produced PfRH5 elicited invasion-blocking antibodies qualitatively comparable to other approaches.
Collapse
Affiliation(s)
- Wesley L. Fotoran
- Department of Parasitology, Institute of Biomedical Sciences,
University of São Paulo, São Paulo, SP, Brazil
| | - Nicole Kleiber
- Department of Parasitology, Institute of Biomedical Sciences,
University of São Paulo, São Paulo, SP, Brazil
- Max Planck Institute of Biophysics, Göttingen, Germany
| | - Thomas Müntefering
- Department of Molecular Physiology, Institute of Animal Physiology,
University of Münster, Münster, Germany
| | - Eva Liebau
- Department of Molecular Physiology, Institute of Animal Physiology,
University of Münster, Münster, Germany
| | - Gerhard Wunderlich
- Department of Parasitology, Institute of Biomedical Sciences,
University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
75
|
Yang S, Arrode-Bruses G, Frank I, Grasperge B, Blanchard J, Gettie A, Martinelli E, Ho EA. Anti-α 4β 7 monoclonal antibody-conjugated nanoparticles block integrin α 4β 7 on intravaginal T cells in rhesus macaques. SCIENCE ADVANCES 2020; 6:6/34/eabb9853. [PMID: 32937372 PMCID: PMC7442472 DOI: 10.1126/sciadv.abb9853] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/10/2020] [Indexed: 06/11/2023]
Abstract
Intravenous administration of anti-α4β7 monoclonal antibody in macaques decreases simian immunodeficiency virus (SIV) vaginal infection and reduces gut SIV loads. Because of potential side effects of systemic administration, a prophylactic strategy based on mucosal administration of anti-α4β7 antibody may be safer and more effective. With this in mind, we developed a novel intravaginal formulation consisting of anti-α4β7 monoclonal antibody-conjugated nanoparticles (NPs) loaded in a 1% hydroxyethylcellulose (HEC) gel (NP-α4β7 gel). When intravaginally administered as a single dose in a rhesus macaque model, the formulation preferentially bound to CD4+ or CD3+ T cells expressing high levels of α4β7, and occupied ~40% of α4β7 expressed by these subsets and ~25% of all cells expressing α4β7 Blocking of the α4β7 was restricted to the vaginal tract without any changes detected systemically.
Collapse
Affiliation(s)
- Sidi Yang
- School of Pharmacy, University of Waterloo, 10 Victoria St. S A, Kitchener, Ontario N2G 1C5, Canada
| | - Geraldine Arrode-Bruses
- Center for Biomedical Research, Population Council, One Dag Hammarskjold Plaza, New York, NY 10017, USA
| | - Ines Frank
- Center for Biomedical Research, Population Council, One Dag Hammarskjold Plaza, New York, NY 10017, USA
| | - Brooke Grasperge
- Tulane National Primate Research Center, Tulane University, 6823 St. Charles Ave., New Orleans, LA 70118, USA
| | - James Blanchard
- Tulane National Primate Research Center, Tulane University, 6823 St. Charles Ave., New Orleans, LA 70118, USA
| | - Agegnehu Gettie
- Aaron Diamond AIDS Research Center, Rockefeller University, 455 1st Avenue #7, New York, NY 10016, USA
| | - Elena Martinelli
- Center for Biomedical Research, Population Council, One Dag Hammarskjold Plaza, New York, NY 10017, USA.
| | - Emmanuel A Ho
- School of Pharmacy, University of Waterloo, 10 Victoria St. S A, Kitchener, Ontario N2G 1C5, Canada.
| |
Collapse
|
76
|
Rossini M, Pizzo P, Filadi R. Better to keep in touch: investigating inter‐organelle cross‐talk. FEBS J 2020; 288:740-755. [DOI: 10.1111/febs.15451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/28/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Michela Rossini
- Department of Biomedical Sciences University of Padua Padua Italy
| | - Paola Pizzo
- Department of Biomedical Sciences University of Padua Padua Italy
- Neuroscience Institute National Research Council (CNR) Padua Italy
| | - Riccardo Filadi
- Department of Biomedical Sciences University of Padua Padua Italy
- Neuroscience Institute National Research Council (CNR) Padua Italy
| |
Collapse
|
77
|
Wu Y, Lian J, Gonçales VR, Pardehkhorram R, Tang W, Tilley RD, Gooding JJ. Patterned Molecular Films of Alkanethiol and PLL-PEG on Gold-Silicate Interfaces: How to Add Functionalities while Retaining Effective Antifouling. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:5243-5250. [PMID: 32323996 DOI: 10.1021/acs.langmuir.0c00586] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Spatial control of surface functionalization and interactions is essential for microarray-based analysis. This study reports the fabrication of two-dimensional molecular films with site-specific functionalities, forming microarrays at discrete locations. Arrays of microsized gold disks were produced on a silicate membrane using microfabrication. On these arrays, orthogonal self-assembly of molecules was performed that can specifically bind to gold or silicate. The gold array elements were functionalized with a range of alkanethiols and the silicate with polymeric poly-l-lysine-grafted-poly(ethylene glycol) (PLL-PEG). The surface functionalization on the gold disk array and the surrounding substrate was characterized at each step using X-ray photoelectron spectroscopy (XPS) to show that alkanethiols are specifically attached to the gold. PLL-PEG was used to provide resistance against nonspecific protein and cell adsorption and attached exclusively to the silicate. The effectiveness of the surface chemistry was validated by the selective self-assembly of a gold nanoparticle monolayer array on the gold regions. In a more sophisticated example, selective adhesion of MCF-7 cells to anti-EpCAM antibody modified gold areas of the gold-silicate surface was demonstrated to give a cell microarray. This study provides a general approach to fabricate chemical patterns on silicon-based devices with the convergence of microfabrication and material-specific surface modification, which may be useful to expand the functionalities and potential applications for patterned biomolecular films. Importantly, the ability to pattern surfaces with different surface chemistries is not limited to planar surfaces using this orthogonal surface-coupling approach.
Collapse
Affiliation(s)
- Yanfang Wu
- School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Jiaxin Lian
- School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Vinicius R Gonçales
- School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Raheleh Pardehkhorram
- School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Wenxian Tang
- School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Richard D Tilley
- School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
- Electron Microscope Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - J Justin Gooding
- School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
78
|
Gessner I, Neundorf I. Nanoparticles Modified with Cell-Penetrating Peptides: Conjugation Mechanisms, Physicochemical Properties, and Application in Cancer Diagnosis and Therapy. Int J Mol Sci 2020; 21:E2536. [PMID: 32268473 PMCID: PMC7177461 DOI: 10.3390/ijms21072536] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/28/2020] [Accepted: 04/03/2020] [Indexed: 12/27/2022] Open
Abstract
Based on their tunable physicochemical properties and the possibility of producing cell-specific platforms through surface modification with functional biomolecules, nanoparticles (NPs) represent highly promising tools for biomedical applications. To improve their potential under physiological conditions and to enhance their cellular uptake, combinations with cell-penetrating peptides (CPPs) represent a valuable strategy. CPPs are often cationic peptide sequences that are able to translocate across biological membranes and to carry attached cargos inside cells and have thus been recognized as versatile tools for drug delivery. Nevertheless, the conjugation of CPP to NP surfaces is dependent on many properties from both individual components, and further insight into this complex interplay is needed to allow for the fabrication of highly stable but functional vectors. Since CPPs per se are nonselective and enter nearly all cells likewise, additional decoration of NPs with homing devices, such as tumor-homing peptides, enables the design of multifunctional platforms for the targeted delivery of chemotherapeutic drugs. In this review, we have updated the recent advances in the field of CPP-NPs, focusing on synthesis strategies, elucidating the influence of different physicochemical properties, as well as their application in cancer research.
Collapse
Affiliation(s)
- Isabel Gessner
- Department of Chemistry, Inorganic Chemistry, University of Cologne, Greinstr 6, 50939 Cologne, Germany;
| | - Ines Neundorf
- Department of Chemistry, Biochemistry, University of Cologne, Zuelpicher Str. 47a, 50674 Cologne, Germany
| |
Collapse
|
79
|
Vaughan HJ, Green JJ, Tzeng SY. Cancer-Targeting Nanoparticles for Combinatorial Nucleic Acid Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901081. [PMID: 31222852 PMCID: PMC6923623 DOI: 10.1002/adma.201901081] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/13/2019] [Indexed: 05/03/2023]
Abstract
Nucleic acids are a promising type of therapeutic for the treatment of a wide range of conditions, including cancer, but they also pose many delivery challenges. For efficient and safe delivery to cancer cells, nucleic acids must generally be packaged into a vehicle, such as a nanoparticle, that will allow them to be taken up by the target cells and then released in the appropriate cellular compartment to function. As with other types of therapeutics, delivery vehicles for nucleic acids must also be designed to avoid unwanted side effects; thus, the ability of such carriers to target their cargo to cancer cells is crucial. Classes of nucleic acids, hurdles that must be overcome for effective intracellular delivery, types of nonviral nanomaterials used as delivery vehicles, and the different strategies that can be employed to target nucleic acid delivery specifically to tumor cells are discussed. Additonally, nanoparticle designs that facilitate multiplexed delivery of combinations of nucleic acids are reviewed.
Collapse
Affiliation(s)
- Hannah J Vaughan
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| |
Collapse
|
80
|
Combined Mini-Parasep SF and Nanogold Immunoassay Show Potential in Stool Antigen Immunodetection for Giardiasis Diagnosis. Sci Rep 2020; 10:2. [PMID: 32225166 PMCID: PMC7103579 DOI: 10.1038/s41598-019-55492-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/27/2019] [Indexed: 12/02/2022] Open
Abstract
Covalent loading or directional binding of biomolecules on gold nanoparticles (AuNPs) could lead to better results than simple direct adsorption for an enhanced ELISA application. The use of Mini-Parasep solvent-free (SF) without ether or ethyl acetate for the clean and efficient concentration of protozoa cysts, it is a single-use device for in vitro diagnostic use only. In this work, we used Mini-Parasep SF for the detection of giardia cysts in comparison to direct smear and Merthiolate-Iodine Formaldehyde Concentration (MIFC) technique in addition to its use in antigen detection by AuNPs biomolecule loading using rabbit polyclonal antibodies (pAb) against purified Giardia antigen (PGA). As a result, Mini-Parasep SF was the most effective method for Giardia cyst detection and regarding optimization of Mini-Parasep antigen detection, our data showed increased sensitivity and specificity of nano-sandwich ELISA to 92% and 94% respectively and increased positive predictive value (PPV) and negative predictive value (NPV) to 88.64% and 95.91% respectively. In conclusion, this research provides that Mini-Parasep SF concentrator enhanced Giardia cyst detection and improved antigen preparation for AuNPs sandwich ELISA in giardiasis diagnosis. The advantages of this method are the short assay time and the raised accuracy of antigen detection providing concentrated samples without the risk of solvent use and being a disposable Mini-Parasep it helps in giardia antigen purification as well as raising the sensitivity and specificity of ELISA through binding AuNPs.
Collapse
|
81
|
Pollok NE, Rabin C, Walgama CT, Smith L, Richards I, Crooks RM. Electrochemical Detection of NT-proBNP Using a Metalloimmunoassay on a Paper Electrode Platform. ACS Sens 2020; 5:853-860. [PMID: 32154707 DOI: 10.1021/acssensors.0c00167] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this paper, we demonstrate an electrochemical method for detection of the heart failure biomarker, N-terminal prohormone brain natriuretic peptide (NT-proBNP). The approach is based on a paper electrode assembly and a metalloimmunoassay; it is intended for eventual integration into a home-use sensor. Sensing of NT-proBNP relies on the formation of a sandwich immunoassay and electrochemical quantification of silver nanoparticle (AgNP) labels attached to the detection antibodies (Abs). There are four important outcomes reported in this article. First, compared to physisorption of the detection Abs on the AgNP labels, a 27-fold increase in signal is observed when a heterobifunctional cross-linker is used to facilitate this labeling. Second, the assay is selective in that it does not cross-react with other cardiac natriuretic peptides. Third, the assay forms in undiluted human serum (though the electrochemical analysis is carried out in buffer). Finally, and most important, the assay is able to detect NT-proBNP at concentrations between 0.58 and 2.33 nM. This performance approaches the critical NT-proBNP concentration threshold often used by physicians for risk stratification purposes: ∼0.116 nM.
Collapse
Affiliation(s)
- Nicole E. Pollok
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Charlie Rabin
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Charuksha T. Walgama
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Leilani Smith
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Ian Richards
- Interactives Executive Excellence LLC, Austin, Texas 78733, United States
| | - Richard M. Crooks
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| |
Collapse
|
82
|
Jordan T, O'Brien MA, Spatarelu CP, Luke GP. Antibody-Conjugated Barium Titanate Nanoparticles for Cell-Specific Targeting. ACS APPLIED NANO MATERIALS 2020; 3:2636-2646. [PMID: 35873656 PMCID: PMC9307239 DOI: 10.1021/acsanm.0c00019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Barium titanate nanoparticles (BTNPs) are gaining popularity in biomedical research because of their piezoelectricity, nonlinear optical properties, and high biocompatibility. However, the potential of BTNPs is limited by the ability to create stable nanoparticle dispersions in water and physiological media. In this work, we report a method of surface modification of BTNPs based on surface hydroxylation followed by covalent attachment of hydrophilic poly(ethylene glycol) (PEG) polymers. This polymer coating allows for additional modifications such as fluorescent labeling, surface charge tuning, or directional conjugation of IgG antibodies. We demonstrate the conjugation of anti-EGFR antibodies to the BTNP surface and show efficient molecular targeting of the nanoparticles to A431 cells. Overall, the reported modifications aim to expand the BTNP applications in molecular imaging, cancer therapy, or noninvasive neurostimulation.
Collapse
Affiliation(s)
- Tomas Jordan
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Mikaela A O'Brien
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | | | - Geoffrey P Luke
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States; Translational Engineering in Cancer Research Program, Norris Cotton Cancer Center, Lebanon, New Hampshire 03766, United States
| |
Collapse
|
83
|
Intracellular Delivery of Anti-SMC2 Antibodies against Cancer Stem Cells. Pharmaceutics 2020; 12:pharmaceutics12020185. [PMID: 32098204 PMCID: PMC7076674 DOI: 10.3390/pharmaceutics12020185] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 11/17/2022] Open
Abstract
Structural maintenance of chromosomes protein 2 (SMC2) is a central component of the condensin complex involved in DNA supercoiling, an essential process for embryonic stem cell survival. SMC2 over-expression has been related with tumorigenesis and cancer malignancy and its inhibition is regarded as a potential therapeutic strategy even though no drugs are currently available. Here, we propose to inhibit SMC2 by intracellular delivery of specific antibodies against the SMC2 protein. This strategy aims to reduce cancer malignancy by targeting cancer stem cells (CSC), the tumoral subpopulation responsible of tumor recurrence and metastasis. In order to prevent degradation and improve cellular internalization, anti-SMC2 antibodies (Ab-SMC2) were delivered by polymeric micelles (PM) based on Pluronic® F127 amphiphilic polymers. Importantly, scaffolding the Ab-SMC2 onto nanoparticles allowed its cellular internalization and highly increased its efficacy in terms of cytotoxicity and inhibition of tumorsphere formation in MDA-MB-231 and HCT116 breast and colon cancer cell lines, respectively. Moreover, in the case of the HCT116 cell line G1, cell-cycle arrest was also observed. In contrast, no effects from free Ab-SMC2 were detected in any case. Further, combination therapy of anti-SMC2 micelles with paclitaxel (PTX) and 5-Fluorouracil (5-FU) was also explored. For this, PTX and 5-FU were respectively loaded into an anti-SMC2 decorated PM. The efficacy of both encapsulated drugs was higher than their free forms in both the HCT116 and MDA-MB-231 cell lines. Remarkably, micelles loaded with Ab-SMC2 and PTX showed the highest efficacy in terms of inhibition of tumorsphere formation in HCT116 cells. Accordingly, our data clearly suggest an effective intracellular release of antibodies targeting SMC2 in these cell models and, further, strong cytotoxicity against CSC, alone and in combined treatments with Standard-of-Care drugs.
Collapse
|
84
|
Sudhakara Prasad K, Cao X, Gao N, Jin Q, Sanjay ST, Henao-Pabon G, Li X. A Low-Cost Nanomaterial-based Electrochemical Immunosensor on Paper for High-Sensitivity Early Detection of Pancreatic Cancer. SENSORS AND ACTUATORS. B, CHEMICAL 2020; 305:127516. [PMID: 32863588 PMCID: PMC7453835 DOI: 10.1016/j.snb.2019.127516] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Due to the lack of specific early detection methods for pancreatic cancer, it usually goes undetected until it is advanced. By employing paper-based electrodes (PPE), herein we for the first time developed a disposable low-cost paper-based immunosensor for rapid early quantitative detection of pancreatic cancer with a new biomarker, pseudopodium-enriched atypical kinase one, SGK269 (PEAK1). The immunosensor was constructed by fabricating PPEs immobilized with the versatile nanomaterial graphene oxide for the incorporation of antibodies to form an immunosensing platform, without the need of complicated surface modification. After it was confirmed that the PPEs exhibited excellent electrochemical properties, a sandwich-type electrochemical immunosensor was subsequently constructed by employing graphene oxide layers immobilized with anti-PEAK1, and the antibody conjugated with gold nanoparticles (AuNPs-tagged-Anti PEAK1). Further, spectral and surface characteristic studies confirmed the formation of the immunosensing platform. The immunosensor for PEAK1 exhibited a wide linear range between 10 pg mL-1 and 106 pg mL-1 with a low limit of detection (LOD) of 10 pg mL-1. The obtained results point towards rapid, sensitive, and specific early diagnosis of pancreatic cancer at the point of care and other low-resource settings.
Collapse
Affiliation(s)
- K. Sudhakara Prasad
- Department of Chemistry & Biochemistry, University of Texas at El Paso, Texas, USA
| | - Xiyue Cao
- Department of Chemistry & Biochemistry, University of Texas at El Paso, Texas, USA
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, PR China
| | - Ning Gao
- Department of Chemistry & Biochemistry, University of Texas at El Paso, Texas, USA
| | - Qijie Jin
- Department of Chemistry & Biochemistry, University of Texas at El Paso, Texas, USA
- College of Materials Science and Engineering, Nanjing Tech University, Nanjing 210009, PR China
| | - Sharma T. Sanjay
- Department of Chemistry & Biochemistry, University of Texas at El Paso, Texas, USA
| | - Gilberto Henao-Pabon
- Biomedical Engineering, Border Biomedical Research Center, and Environmental Science & Engineering, University of Texas at El Paso, USA
| | - XiuJun Li
- Department of Chemistry & Biochemistry, University of Texas at El Paso, Texas, USA
- Biomedical Engineering, Border Biomedical Research Center, and Environmental Science & Engineering, University of Texas at El Paso, USA
- Corresponding Author: XiuJun Li,
| |
Collapse
|
85
|
Huang Z, Wang C, Liu R, Su Y, Lv Y. Self-Validated Homogeneous Immunoassay by Single Nanoparticle in-Depth Scrutinization. Anal Chem 2020; 92:2876-2881. [PMID: 31910615 DOI: 10.1021/acs.analchem.9b05596] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The most convenient method for the clinical routine analysis of disease biomarkers is homogeneous immunoassay, which minimizes the requirements for automation and time-/lab-consumption. Despite great success, because sample constituents are not removed by a separation or washing step, a major challenge in conducting homogeneous immunoassays for the practical application is the matrix effect-related inaccuracy. Herein, to guarantee an accurate quantification, a self-validated homogeneous immunoassay was proposed, by simultaneously scrutinizing both frequency and intensity of single gold nanoparticles. The two analytical modes of single particle inductively coupled plasma mass spectrometry (ICPMS) correlated well with each other, resulting in a self-validation mechanism for the accurate immunoassay. Both two modes of the proposed method provided linear ranges of 2 orders of magnitude and LODs of pM level. Thanks to the self-validated strategy and the high tolerance of the matrix effect of ICPMS, the proposed homogeneous immunoassay was successfully demonstrated in a series of human serum samples, with results in good accordance with clinical routine methods.
Collapse
Affiliation(s)
- Zili Huang
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu , Sichuan 610064 , China
| | - Chaoqun Wang
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu , Sichuan 610064 , China
| | - Rui Liu
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu , Sichuan 610064 , China
| | - Yingying Su
- Analytical & Testing Center , Sichuan University , Chengdu 610064 , China
| | - Yi Lv
- Analytical & Testing Center , Sichuan University , Chengdu 610064 , China
| |
Collapse
|
86
|
Hybrid Particle Swarm Optimization with Sine Cosine Algorithm and Nelder–Mead Simplex for Solving Engineering Design Problems. ARABIAN JOURNAL FOR SCIENCE AND ENGINEERING 2020. [DOI: 10.1007/s13369-019-04285-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
87
|
Antibody-Targeted Nanoparticles for Cancer Treatment. Nanobiomedicine (Rij) 2020. [DOI: 10.1007/978-981-32-9898-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
88
|
Fang H, Li X, Leng Y, Huang X, Xiong Y. Amphiphilic ligand modified gold nanocarriers to amplify lanthanide loading for ultrasensitive DELFIA detection of Cronobacter. Analyst 2020; 145:249-256. [DOI: 10.1039/c9an01945f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Thiolated ethylenediaminetetraacetic acid and thiolated acylhydrazine-terminated ligands modified gold nanoflowers as amplified nanocarriers to increase the Ln3+labeling ratio for improving the sensitivity of traditional DELFIA.
Collapse
Affiliation(s)
- Hao Fang
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang 330047
- P. R. China
- School of Food Science and Technology
| | - Xiangmin Li
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang 330047
- P. R. China
- School of Food Science and Technology
| | - Yuankui Leng
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang 330047
- P. R. China
- School of Food Science and Technology
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang 330047
- P. R. China
- School of Food Science and Technology
| | - Yonghua Xiong
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang 330047
- P. R. China
- School of Food Science and Technology
| |
Collapse
|
89
|
Pollok NE, Rabin C, Smith L, Crooks RM. Orientation-Controlled Bioconjugation of Antibodies to Silver Nanoparticles. Bioconjug Chem 2019; 30:3078-3086. [PMID: 31730333 PMCID: PMC6920564 DOI: 10.1021/acs.bioconjchem.9b00737] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Here we report on the use of heterobifunctional cross-linkers (HBCLs) to control the number, orientation, and activity of immunoglobulin G antibodies (Abs) conjugated to silver nanoparticles (AgNPs). A hydrazone conjugation method resulted in exclusive modification of the polysaccharide chains present on the fragment crystallizable region of the Abs, leaving the antigen-binding regions accessible. Two HBCLs, each having a hydrazide terminal group, were synthesized and tested for effectiveness. The two HBCLs differed in two respects, however: (1) either a thiol or a dithiolane group was used for attachment to the AgNP; and (2) the spacer arm was either a PEG chain or an alkyl chain. Both cross-linkers immobilized 5 ± 1 Abs on the surface of each 20-nm-diameter AgNP. Electrochemical results, obtained using a half-metalloimmunoassay, proved that Abs conjugated to AgNPs via either of the two HBCLs were 4 times more active than those conjugated by the more common physisorption technique. This finding confirmed that the HBCLs exerted orientational control over the Abs. We also demonstrated that the AgNP-HBCL-Ab conjugates were stable and active for at least 2 weeks. Finally, we found that the stability of the HBCLs themselves was related to the nature of their spacer arms. Specifically, the results showed that the HBCL having the alkyl chain is chemically stable for at least 90 days, making it the preferred cross-linker for bioassays.
Collapse
Affiliation(s)
| | | | - Leilani Smith
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th St., Stop A1590, Austin, TX, 78712-1224, U.S.A
| | - Richard M. Crooks
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th St., Stop A1590, Austin, TX, 78712-1224, U.S.A
| |
Collapse
|
90
|
Changalvaie B, Han S, Moaseri E, Scaletti F, Truong L, Caplan R, Cao A, Bouchard R, Truskett TM, Sokolov KV, Johnston KP. Indocyanine Green J Aggregates in Polymersomes for Near-Infrared Photoacoustic Imaging. ACS APPLIED MATERIALS & INTERFACES 2019; 11:46437-46450. [PMID: 31804795 DOI: 10.1021/acsami.9b14519] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Clinical translation of photoacoustic imaging (PAI) has been limited by the lack of near-infrared (NIR) contrast agents with low toxicity required for regulatory approval. Herein, J aggregates of indocyanine green (ICG) with strong NIR absorbance were encapsulated at high loadings within small 77 nm polymersomes (nanocapsules) composed of poly(lactide-co-glycolide-b-poly(ethylene glycol)) (PLGA-b-PEG) bilayers, thus enabling PAI of of breast and ovarian cancer cells with high specificity and a sensitivity at the level of ∼100 total cells. All of the major components of the polymersomes are FDA approved and used in the clinic. During formation of polymersomes with a water-in-oil-in-water double emulsion process, loss of ICG from the ICG J aggregates was minimized by coating them with a layer of branched polyethylenimine and by providing excess "sacrificial" ICG to adsorb at the oil-water interfaces. The encapsulated J aggregates were protected against dissociation by the polymersome shell for 24 h in 100% fetal bovine serum, after which the polymersomes biodegraded and the J aggregates dissociated to ICG monomers.
Collapse
Affiliation(s)
| | - Sangheon Han
- Department of Imaging Physics , MD Anderson Cancer Center , Houston , Texas 77030 , United States
- Department of Bioengineering , Rice University , Houston , Texas 77005 , United States
| | | | | | | | | | | | - Richard Bouchard
- Department of Imaging Physics , MD Anderson Cancer Center , Houston , Texas 77030 , United States
- Graduate School of Biomedical Sciences , The University of Texas MD Anderson Cancer Center , Houston , Texas 77030 , United States
| | | | - Konstantin V Sokolov
- Department of Imaging Physics , MD Anderson Cancer Center , Houston , Texas 77030 , United States
- Department of Bioengineering , Rice University , Houston , Texas 77005 , United States
| | | |
Collapse
|
91
|
Mangini V, Maggi V, Trianni A, Melle F, De Luca E, Pennetta A, Del Sole R, Ventura G, Cataldi TRI, Fiammengo R. Directional Immobilization of Proteins on Gold Nanoparticles Is Essential for Their Biological Activity: Leptin as a Case Study. Bioconjug Chem 2019; 31:74-81. [PMID: 31851492 DOI: 10.1021/acs.bioconjchem.9b00748] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Gold nanomaterials hold great potential for biomedical applications. While this field is evolving rapidly, little attention has been paid to precise nanoparticle design and functionalization. Here, we show that when using proteins as targeting moieties, it is fundamental to immobilize them directionally to preserve their biological activity. Using full-length leptin as a case study, we have developed two alternative conjugation strategies for protein immobilization based on either a site-selective or a nonselective derivatization approach. We show that only nanoparticles with leptin immobilized site-selectively fully retain the ability to interact with the cognate leptin receptor. These results demonstrate the importance of a specified molecular design when preparing nanoparticles labeled with proteins.
Collapse
Affiliation(s)
- Vincenzo Mangini
- Center for Biomolecular Nanotechnologies@UniLe , Istituto Italiano di Tecnologia (IIT) , Via Barsanti , 73010 Arnesano, Lecce , Italy
| | - Vito Maggi
- Center for Biomolecular Nanotechnologies@UniLe , Istituto Italiano di Tecnologia (IIT) , Via Barsanti , 73010 Arnesano, Lecce , Italy.,Dipartimento di Ingegneria dell'Innovazione , Università del Salento , Via per Monteroni Km 1 , 73100 Lecce , Italy
| | - Alberta Trianni
- Center for Biomolecular Nanotechnologies@UniLe , Istituto Italiano di Tecnologia (IIT) , Via Barsanti , 73010 Arnesano, Lecce , Italy
| | - Francesca Melle
- Center for Biomolecular Nanotechnologies@UniLe , Istituto Italiano di Tecnologia (IIT) , Via Barsanti , 73010 Arnesano, Lecce , Italy
| | - Elisa De Luca
- Center for Biomolecular Nanotechnologies@UniLe , Istituto Italiano di Tecnologia (IIT) , Via Barsanti , 73010 Arnesano, Lecce , Italy
| | - Antonio Pennetta
- Dipartimento di Ingegneria dell'Innovazione , Università del Salento , Via per Monteroni Km 1 , 73100 Lecce , Italy.,Dipartimento di Beni Culturali , Università del Salento , Via Dalmazio Birago 64 , 73100 Lecce , Italy
| | - Roberta Del Sole
- Dipartimento di Ingegneria dell'Innovazione , Università del Salento , Via per Monteroni Km 1 , 73100 Lecce , Italy
| | - Giovanni Ventura
- Dipartimento di Chimica , Università degli Studi di Bari Aldo Moro , via Orabona 4 , 70126 Bari , Italy
| | - Tommaso R I Cataldi
- Dipartimento di Chimica , Università degli Studi di Bari Aldo Moro , via Orabona 4 , 70126 Bari , Italy.,Centro Interdipartimentale SMART , Università degli Studi di Bari Aldo Moro , via Orabona 4 , 70126 Bari , Italy
| | - Roberto Fiammengo
- Center for Biomolecular Nanotechnologies@UniLe , Istituto Italiano di Tecnologia (IIT) , Via Barsanti , 73010 Arnesano, Lecce , Italy
| |
Collapse
|
92
|
Luo K, Ryu J, Seol IH, Jeong KB, You SM, Kim YR. Paper-Based Radial Chromatographic Immunoassay for the Detection of Pathogenic Bacteria in Milk. ACS APPLIED MATERIALS & INTERFACES 2019; 11:46472-46478. [PMID: 31746586 DOI: 10.1021/acsami.9b16075] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Here, a paper-based radial flow chromatographic immunoassay (RFCI) employing gold nanoparticles (AuNPs) as chromatic agents was developed for the detection of Escherichia coli O157:H7 in whole milk. A 4-repeated gold-binding peptide-tagged (4GBP) streptococcal protein G (SPG) fusion protein was constructed as a bifunctional linker to immobilize antibodies on the surface of AuNPs with a well-oriented form based on the specific affinity of GBP and SPG to the gold and Fc portion of the antibody, respectively. 4GS@AuNPs prepared with the bifunctional linker protein exhibited excellent colloidal stability even at high salt concentrations of up to 500 mM, which is a critical requirement for its application to a broad range of biological and food samples. The enhanced colloidal stability and excellent binding capability of the immuno-4GS@AuNPs toward target bacteria lowered the detection limit of RFCI for target pathogenic bacteria in whole milk as low as 103 CFU/mL, which is by an order of magnitude lower than that of conventional immuno-AuNPs prepared with physical adsorption of antibodies. The RFCI pattern could also be converted into a grayscale value by simple image processing for quantitative determination of target pathogenic bacteria. This paper-based detection system would provide an effective means of monitoring the presence of food-borne pathogens in real food samples with naked eyes.
Collapse
Affiliation(s)
- Ke Luo
- Graduate School of Biotechnology & Department of Food Science and Biotechnology , Kyung Hee University , Yongin 17104 , Korea
| | - Jian Ryu
- Graduate School of Biotechnology & Department of Food Science and Biotechnology , Kyung Hee University , Yongin 17104 , Korea
| | - In-Hye Seol
- Graduate School of Biotechnology & Department of Food Science and Biotechnology , Kyung Hee University , Yongin 17104 , Korea
| | - Ki-Baek Jeong
- Graduate School of Biotechnology & Department of Food Science and Biotechnology , Kyung Hee University , Yongin 17104 , Korea
| | - Sang-Mook You
- Graduate School of Biotechnology & Department of Food Science and Biotechnology , Kyung Hee University , Yongin 17104 , Korea
| | - Young-Rok Kim
- Graduate School of Biotechnology & Department of Food Science and Biotechnology , Kyung Hee University , Yongin 17104 , Korea
| |
Collapse
|
93
|
Molavipordanjani S, Hosseinimehr SJ. Strategies for Conjugation of Biomolecules to Nanoparticles as Tumor Targeting Agents. Curr Pharm Des 2019; 25:3917-3926. [DOI: 10.2174/1381612825666190903154847] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022]
Abstract
Combination of nanotechnology, biochemistry, chemistry and biotechnology provides the opportunity
to design unique nanoparticles for tumor targeting, drug delivery, medical imaging and biosensing. Nanoparticles
conjugated with biomolecules such as antibodies, peptides, vitamins and aptamer can resolve current challenges
including low accumulation, internalization and retention at the target site in cancer diagnosis and therapy
through active targeting. In this review, we focus on different strategies for conjugation of biomolecules to
nanoparticles such as inorganic nanoparticles (iron oxide, gold, silica and carbon nanoparticles), liposomes, lipid
and polymeric nanoparticles and their application in tumor targeting.
Collapse
Affiliation(s)
- Sajjad Molavipordanjani
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
94
|
Highly sensitive and selective detection of single-nucleotide polymorphisms using gold nanoparticle MutS enzymes and a micro cantilever resonator. Talanta 2019; 205:120154. [DOI: 10.1016/j.talanta.2019.120154] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 07/12/2019] [Accepted: 07/13/2019] [Indexed: 12/21/2022]
|
95
|
Adibnia V, Mirbagheri M, Latreille PL, Faivre J, Cécyre B, Robert J, Bouchard JF, Martinez VA, Delair T, David L, Hwang DK, Banquy X. Chitosan hydrogel micro-bio-devices with complex capillary patterns via reactive-diffusive self-assembly. Acta Biomater 2019; 99:211-219. [PMID: 31473363 DOI: 10.1016/j.actbio.2019.08.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023]
Abstract
We present chitosan hydrogel microfluidic devices with self-assembled complex microcapillary patterns, conveniently formed by a diffusion-reaction process. These patterns in chitosan hydrogels are formed by a single-step procedure involving diffusion of a gelation agent into the polymer solution inside a microfluidic channel. By changing the channel geometry, it is demonstrated how to control capillary length, trajectory and branching. Diffusion of nanoparticles (NPs) in the capillary network is used as a model to effectively mimic the transport of nano-objects in vascularized tissues. Gold NPs diffusion is measured locally in the hydrogel chips, and during their two-step transport through the capillaries to the gel matrix and eventually to embedded cell clusters in the gel. In addition, the quantitative analyses reported in this study provide novel opportunities for theoretical investigation of capillary formation and propagation during diffusive gelation of biopolymers. STATEMENT OF SIGNIFICANCE: Hydrogel micropatterning is a challenging task, which is of interest in several biomedical applications. Creating the patterns through self assembly is highly beneficial, because of the accessible and practical preparation procedure. In this study, we introduced complex self-assembled capillary patterns in chitosan hydrogels using a microfluidic approach. To demonstrate the potential application of these capillary patterns, a vascularized hydrogel with microwells occupied by cells was produced, and the diffusion of gold nanoparticles travelling in the capillaries and diffusing in the gel were evaluated. This model mimics a simplified biological tissue, where nanomedicine has to travel through the vasculature, extravasate into and diffuse through the extracellular matrix and eventually reach targeted cells.
Collapse
|
96
|
Al-Sharqi A, Apun K, Vincent M, Kanakaraju D, Bilung LM, Sum MSH. Investigation of the antibacterial activity of Ag-NPs conjugated with a specific antibody against Staphylococcus aureus after photoactivation. J Appl Microbiol 2019; 128:102-115. [PMID: 31596989 DOI: 10.1111/jam.14471] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 09/11/2019] [Accepted: 09/26/2019] [Indexed: 11/26/2022]
Abstract
AIM This work reports a new method for the use of lasers for the selective killing of bacteria targeted using light-absorbing Silver nanoparticles (Ag-NPs) conjugated with a specific antibody against the Gram-positive bacterium Staphylococcus aureus (S. aureus). METHODS AND RESULTS Ag-NPs were synthesized using a chemical reduction method and characterized with respect to their surface plasmon resonance, surface morphology via transmission electron microscopy (TEM) and dynamic light scattering (DLS). The bacterial surface was targeted using 20 nm Ag-NPs conjugated with an anti-protein A antibody. Labelled bacteria were irradiated with blue visible laser at 2·04 W/cm2 . The antibacterial activity of functionalized Ag-NPs was investigated by fluorescence microscopy after irradiation, and morphological changes in S. aureus after laser treatment were assessed using scanning electron microscopy (SEM). The laser-irradiated, functionalized Ag-NPs exhibited significant bactericidal activity, and laser-induced bacterial damage was observed after 10 min of laser irradiation against S. aureus. The fluorescence microscopic analysis results supported that bacterial cell death occurred in the presence of the functionalized Ag-NPs. CONCLUSIONS The results of this study suggest that a novel method for the preparation of functionalized nanoparticles has potential as a potent antibacterial agent for the selective killing of resistant disease-causing bacteria. SIGNIFICANCE AND IMPACT OF THE STUDY This study shows that Ag-NPs functionalized with a specific antibody, could be used in combination with laser radiation as a novel treatment to target resistant bacterial and fungal pathogens with minimal impact on normal microflora.
Collapse
Affiliation(s)
- A Al-Sharqi
- Faculty of Resource Science and Technology, University Malaysia Sarawak, 94300, Kota Samarahan, Sarawak, Malaysia
| | - K Apun
- Faculty of Resource Science and Technology, University Malaysia Sarawak, 94300, Kota Samarahan, Sarawak, Malaysia
| | - M Vincent
- Faculty of Resource Science and Technology, University Malaysia Sarawak, 94300, Kota Samarahan, Sarawak, Malaysia
| | - D Kanakaraju
- Faculty of Resource Science and Technology, University Malaysia Sarawak, 94300, Kota Samarahan, Sarawak, Malaysia
| | | | - M S H Sum
- Institute of Health & Community Medicine, University Malaysia Sarawak, 94300, Kota Samarahan, Sarawak, Malaysia
| |
Collapse
|
97
|
Shahrabi Farahani M, Mohsenzadegan M, Taeb J, Farajollahi MM. In-vitro prostate cancer biomarker detection by directed conjugation of anti-PSCA antibody to super paramagnetic iron oxide nanoparticless. Med J Islam Repub Iran 2019; 33:16. [PMID: 31380306 PMCID: PMC6662544 DOI: 10.34171/mjiri.33.16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Indexed: 02/06/2023] Open
Abstract
Background: The main property of a successful conjugation of antibodies to nanoparticles is keeping the potency of antibody for binding the antigen, and an oriented conjugation can do that. Under such ground, this study was carried out to explore the efficiency of two conjugation methods in binding iron nanoparticles to an antibody produced against PSCA (prostate stem cell antigen) using in vitro labeling of PC3 cells.
Methods: In this experimental study, we conjugated dextran-superparamagnetic iron oxide nanoparticles (dexSPIONs) to anti-PSCA antibody by two different methods, including targeting carbohydrate moieties in FC domain and the free amine group of amino acid side chains. Ultimately, Iron staining was done by anti-PSCA antibody-dexSPIONs in PC3 cells to detect antibody binding to the cells.
Results: A strong blue dye was induced by iron staining in conjugated dexSPIONs on the membrane of PC3 cells by the former method than the second one. Moreover, cells treated with 20 nm diameters of dexSPIONs showed higher resolution of blue color than those treated with 100 nm nanoparticles.
Conclusion: This oriented conjugation method promoted the efficiency of targeting tumor antigens, and the presence of iron particles might enhance MRI image intensity in vivo by targeting PSCA-overexpressing cells in future studies.
Collapse
Affiliation(s)
- Mahboube Shahrabi Farahani
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Monireh Mohsenzadegan
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Jaleh Taeb
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad M Farajollahi
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.,Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
98
|
Wu CY, Lin JJ, Chang WY, Hsieh CY, Wu CC, Chen HS, Hsu HJ, Yang AS, Hsu MH, Kuo WY. Development of theranostic active-targeting boron-containing gold nanoparticles for boron neutron capture therapy (BNCT). Colloids Surf B Biointerfaces 2019; 183:110387. [PMID: 31394419 DOI: 10.1016/j.colsurfb.2019.110387] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/08/2019] [Accepted: 07/23/2019] [Indexed: 12/13/2022]
Abstract
Successful boron neutron capture therapy (BNCT) requires sufficient and specific delivery of boron atoms to malignant cells. Gold nanoparticles (AuNPs) have been used as a useful delivery system for selectively releasing cytotoxic payloads in the tumor. However, studies demonstrating the in vivo distribution or pharmacokinetics of boron-containing AuNPs via noninvasive imaging are lacking. This study aims to develop theranostic AuNP-boron cage assemblies (B-AuNPs) and evaluate its feasibility for BNCT. The commercial citrate-coated AuNPs were subjected to PEGylation, azide addition, and carborane modification on the surface. To further arm the AuNPs, we conjugated anti-HER2 antibody (61 IgG) with boron-containing PEGylated AuNPs to form 61-B-AuNPs. The diameter and radiolabeling efficiency of boron-containing AuNPs were determined by dynamic light scattering (DLS) and radio thin-layer chromatography (radio TLC), respectively. Noninvasive single-photon emission computed tomography (SPECT)/computed tomography (CT) imaging was performed to determine the pharmacokinetics of radioiodinated AuNPs in N87 gastric cancer xenografts, and the content of boron in tumor and muscle was assessed by inductively coupled plasma mass spectrometry (ICP-MS). After the 3-step modification, the diameter of B-AuNPs increased by ˜25 nm, and antibody conjugation did not affect the diameter of AuNPs. Radioactive iodine (I-123) was introduced in AuNPs by Click chemistry under copper catalysis. The radiolabeling efficiency of 123I-B-AuNPs and 123I-61-B-AuNPs was approximately 60 ± 5%. After purification, the radiochemical purity (RCP) of these NPs was greater than 90%. MicroSPECT/CT imaging showed that the tumor-to-muscle (T/M) ratio of 123I-B-AuNP-injected mice reached 1.91 ± 0.17 at 12 h post-injection, while that of 123I-61-B-AuNP-injected mice was 12.02 ± 0.94. However, the increased uptake of AuNPs by the thyroid was observed at 36 h after the administration of 123I-61-B-AuNPs, indicating antibody-mediated phagocytosis. The T/M ratio, assessed by ICP-MS, of B-AuNP- and 61-B-AuNP-injected mice was 4.91 ± 2.75 and 41.05 ± 11.15, respectively. We successfully developed detectable HER2-targeting boron-containing AuNPs with high RCP and an acceptable yield. Noninvasive imaging could be a valuable tool for the noninvasive determination of the pharmacokinetics of AuNPs and measurement of boron concentration in the tumor.
Collapse
Affiliation(s)
- Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan; Master Program for Biomedical Engineering, China Medical University, Taichung, Taiwan.
| | - Jia-Jia Lin
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Wen-Yi Chang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Cheng-Ying Hsieh
- Department of Chemistry, National Changhua University of Education, Changhua, Taiwan
| | - Chin-Ching Wu
- Department of Public Health, China Medical University, Taichung, Taiwan
| | - Hong-Sen Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hung-Ju Hsu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - An-Suei Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ming-Hua Hsu
- Department of Chemistry, National Changhua University of Education, Changhua, Taiwan
| | - Wei-Ying Kuo
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
99
|
Han S, Bouchard R, Sokolov KV. Molecular photoacoustic imaging with ultra-small gold nanoparticles. BIOMEDICAL OPTICS EXPRESS 2019; 10:3472-3483. [PMID: 31360601 PMCID: PMC6640831 DOI: 10.1364/boe.10.003472] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 05/14/2023]
Abstract
Gold nanoparticles (AuNPs) below 10 nm in size can undergo renal clearance, which could facilitate their clinical translation. However, due to non-linear, direct relationship between their absorption and size, use of such "ultra-small" AuNPs as contrast agents for photoacoustic imaging (PAI) is challenging. This problem is complicated by the tendency of absorption for ultra-small AuNPs to be below the NIR range, which is optimal for in vivo imaging. Herein, we present 5-nm molecularly activated plasmonic nanosensors (MAPS) that produce a strong photoacoustic signal in labeled cancer cells in the NIR, demonstrating the feasibility of sensitive PAI with ultra-small AuNPs.
Collapse
Affiliation(s)
- Sangheon Han
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
- Department of Imaging Physics, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Richard Bouchard
- Department of Imaging Physics, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Konstantin V. Sokolov
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
- Department of Imaging Physics, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| |
Collapse
|
100
|
Vukojicic P, Béhar G, Tawara MH, Fernandez-Villamarin M, Pecorari F, Fernandez-Megia E, Mouratou B. Multivalent Affidendrons with High Affinity and Specificity toward Staphylococcus aureus as Versatile Tools for Modulating Multicellular Behaviors. ACS APPLIED MATERIALS & INTERFACES 2019; 11:21391-21398. [PMID: 31120726 DOI: 10.1021/acsami.9b05702] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Multivalency is a widely occurring natural phenomenon often exploited in nanotechnology to enhance biorecognition. We report the preparation and characterization of versatile, multivalent Affitin-dendrimer conjugates (Affidendrons) showcased by a set targeting Staphylococcus aureus ( S. aureus), an opportunistic pathogen causing numerous hospital- and community-acquired infections. Affitins are small affinity proteins characterized by higher stability and lower cost-effective production than antibodies. The strategy presented provides a platform for the rational design of multivalent nanodevices that, retaining the ability of Affitins to recognize their target with high specificity, achieve a largely enhanced affinity. Affidendrons with precisely designed size and valency have been exploited to modulate complex multicellular behaviors of S. aureus, such as agglutination and biofilm formation. Agglutination assays showed that Affidendrons rapidly cross-link S. aureus strains with high bacterial cell selectivity. Moreover, remarkably low concentrations of Affidendrons were able to effectively prevent biofilm formation. Overall, Affidendrons represent a promising platform for the rapid and selective pathogen identification, infection imaging, and theranostic applications.
Collapse
Affiliation(s)
- Petar Vukojicic
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes , Nantes , France
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica , Universidade de Santiago de Compostela , Jenaro de la Fuente s/n , 15782 Santiago de Compostela , Spain
| | - Ghislaine Béhar
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes , Nantes , France
| | - Maun H Tawara
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica , Universidade de Santiago de Compostela , Jenaro de la Fuente s/n , 15782 Santiago de Compostela , Spain
| | - Marcos Fernandez-Villamarin
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica , Universidade de Santiago de Compostela , Jenaro de la Fuente s/n , 15782 Santiago de Compostela , Spain
| | - Frédéric Pecorari
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes , Nantes , France
| | - Eduardo Fernandez-Megia
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica , Universidade de Santiago de Compostela , Jenaro de la Fuente s/n , 15782 Santiago de Compostela , Spain
| | - Barbara Mouratou
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes , Nantes , France
| |
Collapse
|