1
|
Wang Y, Yang J, Amier Y, Yuan D, Xun Y, Yu X. Advancements in Nanomedicine for the Diagnosis and Treatment of Kidney Stones. Int J Nanomedicine 2025; 20:1401-1423. [PMID: 39925679 PMCID: PMC11805677 DOI: 10.2147/ijn.s504318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/17/2025] [Indexed: 02/11/2025] Open
Abstract
Kidney stones constitute a common condition impacting the urinary system. In clinical diagnosis and management, traditional surgical interventions and pharmacological treatments are primarily utilized; however, these methods possess inherent limitations. Presently, the field of nanomedicine is undergoing significant advancements. The application of nanomaterials in biosensors enables the accurate assessment of urinary ion composition. Furthermore, contrast agents developed from these materials can improve the signal-to-noise ratio and enhance image clarity. By mitigating oxidative stress-induced cellular damage, nanomaterials can inhibit the formation of kidney stones and enhance the efficacy of drug delivery as effective carriers. Additionally, by modifying the physical and chemical properties of bacteria, nanomaterials can effectively eliminate bacterial presence, thereby preventing severe complications. This review explores the advancements in nanomaterials technology related to the early detection of risk factors, clinical diagnosis, and treatment of kidney stones and their associated complications.
Collapse
Affiliation(s)
- Yongqi Wang
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Junyi Yang
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yirixiatijiang Amier
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Dongfeng Yuan
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yang Xun
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiao Yu
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
2
|
Kapare H, Bhosale M, Bhole R. Navigating the future: Advancements in monoclonal antibody nanoparticle therapy for cancer. J Drug Deliv Sci Technol 2025; 104:106495. [DOI: 10.1016/j.jddst.2024.106495] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Mariotti M, Giacon N, Lo Cascio E, Cacaci M, Picchietti S, Di Vito M, Sanguinetti M, Arcovito A, Bugli F. Functionalized PLGA-Based Nanoparticles with Anti-HSV-2 Human Monoclonal Antibody: A Proof of Concept for Early Diagnosis and Targeted Therapy. Pharmaceutics 2024; 16:1218. [PMID: 39339254 PMCID: PMC11434782 DOI: 10.3390/pharmaceutics16091218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Functionalized nanoparticles (NPs) represent a cutting edge in innovative clinical approaches, allowing for the delivery of selected compounds with higher specificity in a wider time frame. They also hold promise for novel theranostic applications that integrate both diagnostic and therapeutic functions. Pathogens are continuously evolving to try to escape the strategies designed to treat them. Objectives: In this work, we describe the development of a biotechnological device, Nano-Immuno-Probes (NIPs), for early detection and infections treatment. Human Herpes Simplex Virus 2 was chosen as model pathogen. Methods: NIPs consist of PLGA-PEG-Sulfone polymeric NPs conjugated to recombinant Fab antibody fragments targeting the viral glycoprotein G2. NIPs synthesis involved multiple steps and was validated through several techniques. Results: DLS analysis indicated an expected size increase with a good polydispersity index. Z-average and z-potential values were measured for PLGA-PEG-Bis-Sulfone NPs (86.6 ± 10.9 nm; -0.7 ± 0.3 mV) and NIPs (151 ± 10.4 nm; -5.1 ± 1.9 mV). SPR assays confirmed NIPs' specificity for the glycoprotein G2, with an apparent KD of 1.03 ± 0.61 µM. NIPs exhibited no cytotoxic effects on VERO cells at 24 and 48 h. Conclusions: This in vitro study showed that NIPs effectively target HSV-2, suggesting the potential use of these nanodevices to deliver both contrast agents as well as therapeutic compounds.
Collapse
Affiliation(s)
- Melinda Mariotti
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (M.M.); (N.G.); (E.L.C.); (M.C.); (M.D.V.); (M.S.)
| | - Noah Giacon
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (M.M.); (N.G.); (E.L.C.); (M.C.); (M.D.V.); (M.S.)
| | - Ettore Lo Cascio
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (M.M.); (N.G.); (E.L.C.); (M.C.); (M.D.V.); (M.S.)
| | - Margherita Cacaci
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (M.M.); (N.G.); (E.L.C.); (M.C.); (M.D.V.); (M.S.)
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Simona Picchietti
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, Largo dell’Università snc, 01100 Viterbo, Italy;
| | - Maura Di Vito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (M.M.); (N.G.); (E.L.C.); (M.C.); (M.D.V.); (M.S.)
| | - Maurizio Sanguinetti
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (M.M.); (N.G.); (E.L.C.); (M.C.); (M.D.V.); (M.S.)
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (M.M.); (N.G.); (E.L.C.); (M.C.); (M.D.V.); (M.S.)
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy
| | - Francesca Bugli
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (M.M.); (N.G.); (E.L.C.); (M.C.); (M.D.V.); (M.S.)
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
4
|
Zhang Y, Tian J. Strategies, Challenges, and Prospects of Nanoparticles in Gynecological Malignancies. ACS OMEGA 2024; 9:37459-37504. [PMID: 39281920 PMCID: PMC11391544 DOI: 10.1021/acsomega.4c04573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/18/2024]
Abstract
Gynecologic cancers are a significant health issue for women globally. Early detection and successful treatment of these tumors are crucial for the survival of female patients. Conventional therapies are often ineffective and harsh, particularly in advanced stages, necessitating the exploration of new therapy options. Nanotechnology offers a novel approach to biomedicine. A novel biosensor utilizing bionanotechnology can be employed for early tumor identification and therapy due to the distinctive physical and chemical characteristics of nanoparticles. Nanoparticles have been rapidly applied in the field of gynecologic malignancies, leading to significant advancements in recent years. This study highlights the significance of nanoparticles in treating gynecological cancers. It focuses on using nanoparticles for precise diagnosis and continuous monitoring of the disease, innovative imaging, and analytic methods, as well as multifunctional drug delivery systems and targeted therapies. This review examines several nanocarrier systems, such as dendrimers, liposomes, nanocapsules, and nanomicelles, for gynecological malignancies. The review also examines the enhanced therapeutic potential and targeted delivery of ligand-functionalized nanoformulations for gynecological cancers compared to nonfunctionalized anoformulations. In conclusion, the text also discusses the constraints and future exploration prospects of nanoparticles in chemotherapeutics. Nanotechnology will offer precise methods for diagnosing and treating gynecological cancers.
Collapse
Affiliation(s)
- Yingfeng Zhang
- University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Jing Tian
- University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| |
Collapse
|
5
|
Behnke M, Holick CT, Vollrath A, Schubert S, Schubert US. Knowledge-Based Design of Multifunctional Polymeric Nanoparticles. Handb Exp Pharmacol 2024; 284:3-26. [PMID: 37017790 DOI: 10.1007/164_2023_649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
Conventional drug delivery systems (DDS) today still face several drawbacks and obstacles. High total doses of active pharmaceutical ingredients (API) are often difficult or impossible to deliver due to poor solubility of the API or undesired clearance from the body caused by strong interactions with plasma proteins. In addition, high doses lead to a high overall body burden, in particular if they cannot be delivered specifically to the target site. Therefore, modern DDS must not only be able to deliver a dose into the body, but should also overcome the hurdles mentioned above as examples. One of these promising devices are polymeric nanoparticles, which can encapsulate a wide range of APIs despite having different physicochemical properties. Most importantly, polymeric nanoparticles are tunable to obtain tailored systems for each application. This can already be achieved via the starting material, the polymer, by incorporating, e.g., functional groups. This enables the particle properties to be influenced not only specifically in terms of their interactions with APIs, but also in terms of their general properties such as size, degradability, and surface properties. In particular, the combination of size, shape, and surface modification allows polymeric nanoparticles to be used not only as a simple drug delivery device, but also to achieve targeting. This chapter discusses to what extent polymers can be designed to form defined nanoparticles and how their properties affect their performance.
Collapse
Affiliation(s)
- Mira Behnke
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| | - Caroline T Holick
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| | - Antje Vollrath
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| | - Stephanie Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| | - Ulrich S Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
6
|
Agwa MM, Elmotasem H, Moustafa RI, Abdelsattar AS, Mohy-Eldin MS, Fouda MMG. Advent in proteins, nucleic acids, and biological cell membranes functionalized nanocarriers to accomplish active or homologous tumor targeting for smart amalgamated chemotherapy/photo-therapy: A review. Int J Biol Macromol 2023; 253:127460. [PMID: 37866559 DOI: 10.1016/j.ijbiomac.2023.127460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/13/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
Conventional cancer mono-therapeutic approaches including radiotherapy, surgery, and chemotherapy don't always achieve satisfactory outcomes and are frequently associated with significant limitations. Although chemotherapy is a vital intervention, its effectiveness is frequently inadequate and is associated with metastasis, multidrug resistance, off-target effect, and normal cells toxicity. Phototherapies are employed in cancer therapy, encompassing photo-dynamic and photo-thermal therapies which under favorable NIR laser light irradiation initiate the included photosensitizers and photo-thermal agents to generate ROS or thermal heat respectively for cancer cells destruction. Photo-therapy is considered noninvasive, posing no resistance, but it still suffers from several pitfalls like low penetration depth and excessive heat generation affecting neighboring tissues. Improved selectivity and tumor-homing capacity could be attained through surface modulation of nanoparticles with targeting ligands that bind to receptors, which are exclusively overexpressed on cancerous cells. Developing novel modified targeted nanoparticulate platforms integrating different therapeutic modalities like photo-therapy and chemotherapy is a topic of active research. This review aimed to highlight recent advances in proteins, nucleic acids, and biological cell membranes functionalized nanocarriers for smart combinatorial chemotherapy/photo-therapy. Nanocarriers decorated with precise targeting ligands, like aptamers, antibody, and lactoferrin, to achieve active tumor-targeting or camouflaging using various biological cell membrane coating are designed to achieve homologous tumor-targeting.
Collapse
Affiliation(s)
- Mona M Agwa
- Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Institute, National Research Centre, 33 El- Behooth St., Dokki, Giza 12622, Egypt.
| | - Heba Elmotasem
- Pharmaceutical Technology Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, 33 El- Behooth St., Dokki, Giza 12622, Egypt
| | - Rehab I Moustafa
- Department of Microbial Biotechnology, Biotechnology Research Institute, National Research Centre, Dokki, Giza 12622, Egypt
| | - Abdallah S Abdelsattar
- Center for Microbiology and Phage Therapy, Zewail City of Science and Technology, October Gardens, 6th of October City, Giza 12578, Egypt
| | - Mohamed S Mohy-Eldin
- Polymer Materials Research Department, Advanced Technology and New Materials Research Institute (ATNMRI), City of Scientific Research and Technological Applications (SRTA-City), P.O. Box 21934, New Borg El-Arab City, Alexandria, Egypt
| | - Moustafa M G Fouda
- Pre-Treatment and Finishing of Cellulosic Fabric Department, Textile Research and Technology Institute, (TRT) National Research Centre, 33 El- Behooth St., Dokki, Giza 12622, Egypt.
| |
Collapse
|
7
|
Houlihan I, Kang B, De S, Krishna V. Photonic Lithotripsy: Near-Infrared Laser Activated Nanomaterials for Kidney Stone Comminution. NANO LETTERS 2023; 23:5981-5988. [PMID: 37358929 PMCID: PMC10348310 DOI: 10.1021/acs.nanolett.3c01166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/20/2023] [Indexed: 06/28/2023]
Abstract
Near-infrared activated nanomaterials have been reported for biomedical applications ranging from photothermal tumor destruction to biofilm eradication and energy-gated drug delivery. However, the focus so far has been on soft tissues, and little is known about energy delivery to hard tissues, which have thousand-fold higher mechanical strength. We present photonic lithotripsy with carbon and gold nanomaterials for fragmenting human kidney stones. The efficacy of stone comminution is dependent on the size and photonic properties of the nanomaterials. Surface restructuring and decomposition of calcium oxalate to calcium carbonate support the contribution of photothermal energy to stone failure. Photonic lithotripsy has several advantages over current laser lithotripsy, including low operating power, noncontact laser operation (distances of at least 10 mm), and ability to break all common stones. Our observations can inspire the development of rapid, minimally invasive techniques for kidney stone treatment and extrapolate to other hard tissues such as enamel and bone.
Collapse
Affiliation(s)
- Ian Houlihan
- Biomedical
Engineering, Lerner Research Institute,
Cleveland Clinic, Cleveland Ohio 44195, United States
| | - Benjamin Kang
- Biomedical
Engineering, Lerner Research Institute,
Cleveland Clinic, Cleveland Ohio 44195, United States
| | - Smita De
- Urology, Glickman
Urology and Kidney Institute, Cleveland Clinic, Cleveland Ohio 44195, United States
- Urology,
Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland Ohio 44106, United States
| | - Vijay Krishna
- Biomedical
Engineering, Lerner Research Institute,
Cleveland Clinic, Cleveland Ohio 44195, United States
- Biomedical
Engineering, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland Ohio 44106, United States
| |
Collapse
|
8
|
Lehot V, Neuberg P, Ripoll M, Daubeuf F, Erb S, Dovgan I, Ursuegui S, Cianférani S, Kichler A, Chaubet G, Wagner A. Targeted Anticancer Agent with Original Mode of Action Prepared by Supramolecular Assembly of Antibody Oligonucleotide Conjugates and Cationic Nanoparticles. Pharmaceutics 2023; 15:1643. [PMID: 37376091 DOI: 10.3390/pharmaceutics15061643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
Despite their clinical success, Antibody-Drug Conjugates (ADCs) are still limited to the delivery of a handful of cytotoxic small-molecule payloads. Adaptation of this successful format to the delivery of alternative types of cytotoxic payloads is of high interest in the search for novel anticancer treatments. Herein, we considered that the inherent toxicity of cationic nanoparticles (cNP), which limits their use as oligonucleotide delivery systems, could be turned into an opportunity to access a new family of toxic payloads. We complexed anti-HER2 antibody-oligonucleotide conjugates (AOC) with cytotoxic cationic polydiacetylenic micelles to obtain Antibody-Toxic-Nanoparticles Conjugates (ATNPs) and studied their physicochemical properties, as well as their bioactivity in both in vitro and in vivo HER2 models. After optimising their AOC/cNP ratio, the small (73 nm) HER2-targeting ATNPs were found to selectively kill antigen-positive SKBR-2 cells over antigen-negative MDA-MB-231 cells in serum-containing medium. Further in vivo anti-cancer activity was demonstrated in an SKBR-3 tumour xenograft model in BALB/c mice in which stable 60% tumour regression could be observed just after two injections of 45 pmol of ATNP. These results open interesting prospects in the use of such cationic nanoparticles as payloads for ADC-like strategies.
Collapse
Affiliation(s)
- Victor Lehot
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Patrick Neuberg
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Manon Ripoll
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - François Daubeuf
- UAR3286, Plate-Forme de Chimie Biologique Intégrative de Strasbourg, ESBS, CNRS-Strasbourg University, 67400 Illkirch-Graffenstaden, France
| | - Stéphane Erb
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Institut du Médicament de Strasbourg, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Igor Dovgan
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Sylvain Ursuegui
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Institut du Médicament de Strasbourg, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Antoine Kichler
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Guilhem Chaubet
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Alain Wagner
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
9
|
Kalaiselvan CR, Laha SS, Somvanshi SB, Tabish TA, Thorat ND, Sahu NK. Manganese ferrite (MnFe2O4) nanostructures for cancer theranostics. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
10
|
Aghajanzadeh M, Zamani M, Rajabi Kouchi F, Eixenberger J, Shirini D, Estrada D, Shirini F. Synergic Antitumor Effect of Photodynamic Therapy and Chemotherapy Mediated by Nano Drug Delivery Systems. Pharmaceutics 2022; 14:pharmaceutics14020322. [PMID: 35214054 PMCID: PMC8880656 DOI: 10.3390/pharmaceutics14020322] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
This review provides a summary of recent progress in the development of different nano-platforms for the efficient synergistic effect between photodynamic therapy and chemotherapy. In particular, this review focuses on various methods in which photosensitizers and chemotherapeutic agents are co-delivered to the targeted tumor site. In many cases, the photosensitizers act as drug carriers, but this review, also covers different types of appropriate nanocarriers that aid in the delivery of photosensitizers to the tumor site. These nanocarriers include transition metal, silica and graphene-based materials, liposomes, dendrimers, polymers, metal–organic frameworks, nano emulsions, and biologically derived nanocarriers. Many studies have demonstrated various benefits from using these nanocarriers including enhanced water solubility, stability, longer circulation times, and higher accumulation of therapeutic agents/photosensitizers at tumor sites. This review also describes novel approaches from different research groups that utilize various targeting strategies to increase treatment efficacy through simultaneous photodynamic therapy and chemotherapy.
Collapse
Affiliation(s)
- Mozhgan Aghajanzadeh
- Department of Chemistry, College of Science, University of Guilan, Rasht 41335-19141, Iran; (M.A.); (M.Z.)
| | - Mostafa Zamani
- Department of Chemistry, College of Science, University of Guilan, Rasht 41335-19141, Iran; (M.A.); (M.Z.)
| | - Fereshteh Rajabi Kouchi
- Micron School of Materials Science and Engineering, Boise State University, Boise, ID 83725, USA; (F.R.K.); (D.E.)
| | - Josh Eixenberger
- Micron School of Materials Science and Engineering, Boise State University, Boise, ID 83725, USA; (F.R.K.); (D.E.)
- Center for Advanced Energy Studies, Boise State University, Boise, ID 83725, USA
- Correspondence: (J.E.); or (F.S.)
| | - Dorsa Shirini
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran;
| | - David Estrada
- Micron School of Materials Science and Engineering, Boise State University, Boise, ID 83725, USA; (F.R.K.); (D.E.)
- Center for Advanced Energy Studies, Boise State University, Boise, ID 83725, USA
| | - Farhad Shirini
- Department of Chemistry, College of Science, University of Guilan, Rasht 41335-19141, Iran; (M.A.); (M.Z.)
- Correspondence: (J.E.); or (F.S.)
| |
Collapse
|
11
|
Zumaya ALV, Rimpelová S, Štějdířová M, Ulbrich P, Vilčáková J, Hassouna F. Antibody Conjugated PLGA Nanocarriers and Superparmagnetic Nanoparticles for Targeted Delivery of Oxaliplatin to Cells from Colorectal Carcinoma. Int J Mol Sci 2022; 23:ijms23031200. [PMID: 35163122 PMCID: PMC8835878 DOI: 10.3390/ijms23031200] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 02/04/2023] Open
Abstract
Anti-CD133 monoclonal antibody (Ab)-conjugated poly(lactide-co-glycolide) (PLGA) nanocarriers, for the targeted delivery of oxaliplatin (OXA) and superparamagnetic nanoparticles (IO-OA) to colorectal cancer cells (CaCo-2), were designed, synthesized, characterized, and evaluated in this study. The co-encapsulation of OXA and IO-OA was achieved in two types of polymeric carriers, namely, PLGA and poly(lactide-co-glycolide)-poly(ethylene glycol) (PLGA-PEG) by double emulsion. PLGA_IO-OA_OXA and PEGylated PLGA_IO-OA_OXA nanoparticles displayed a comparable mean diameter of 207 ± 70 nm and 185 ± 119 nm, respectively. The concentration of the released OXA from the PEGylated PLGA_IO-OA_OXA increased very rapidly, reaching ~100% release after only 2 h, while the PLGA_IO-OA_OXA displayed a slower and sustained drug release. Therefore, for a controlled OXA release, non-PEGylated PLGA nanoparticles were more convenient. Interestingly, preservation of the superparamagnetic behavior of the IO-OA, without magnetic hysteresis all along the dissolution process, was observed. The non-PEGylated nanoparticles (PLGA_OXA, PLGA_IO-OA_OXA) were selected for the anti-CD133 Ab conjugation. The affinity of Ab-coated nanoparticles for CD133-positive cells was examined using fluorescence microscopy in CaCo-2 cells, which was followed by a viability assay.
Collapse
Affiliation(s)
- Alma Lucia Villela Zumaya
- Faculty of Chemical Engineering, University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic; (A.L.V.Z.); (M.Š.)
| | - Silvie Rimpelová
- Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic; (S.R.); (P.U.)
| | - Markéta Štějdířová
- Faculty of Chemical Engineering, University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic; (A.L.V.Z.); (M.Š.)
| | - Pavel Ulbrich
- Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic; (S.R.); (P.U.)
| | - Jarmila Vilčáková
- Faculty of Technology, Tomas Bata University, 760 01 Zlín, Czech Republic;
| | - Fatima Hassouna
- Faculty of Chemical Engineering, University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic; (A.L.V.Z.); (M.Š.)
- Correspondence: ; Tel.: +420-220-444-099
| |
Collapse
|
12
|
Francis DM, Manspeaker MP, Archer PA, Sestito LF, Heiler AJ, Schudel A, Thomas SN. Drug-eluting immune checkpoint blockade antibody-nanoparticle conjugate enhances locoregional and systemic combination cancer immunotherapy through T lymphocyte targeting. Biomaterials 2021; 279:121184. [PMID: 34678650 PMCID: PMC8639654 DOI: 10.1016/j.biomaterials.2021.121184] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 09/03/2021] [Accepted: 10/09/2021] [Indexed: 11/21/2022]
Abstract
Multiple small molecule immune modulators have been identified as synergistic with immune checkpoint blockade (ICB) in their effects on T lymphocytes, but are limited in their successful application to combination cancer immunotherapy due to their short in vivo retention and lack of affinity for T cells. We engineered an antibody-nanoparticle conjugate (ANC) platform consisting of 30 nm polymer nanoparticles that, due to their size and formulation, efficiently distribute after administration to lymph nodes, tissues highly enriched in lymphocytes that contribute to tumor control mediated by ICB. Displaying monoclonal antibodies against surface-expressed T cell markers, NP delivery in vivo to circulating and lymph node-resident lymphocytes was substantially enhanced, as was delivery of small molecules formulated into the NP by passive encapsulation. Using ICB monoclonal antibodies as both targeting moiety and signal-blocking therapeutic, ANCs improved the local and systemic anti-tumor effects of small molecule TGFβ receptor 1 inhibitor and an adenosine 2A antagonist when administered either locoregionally or systemically into the circulation in two syngeneic, aggressive tumor models, slowing tumor growth and prolonging animal survival. As these benefits were lost in the absence of ANC targeting, co-formulation strategies enabling the targeted co-delivery of multiple immunotherapeutics to T lymphocytes have high potential to improve ICB cancer immunotherapy by concurrent inhibition of non-redundant suppressive pathways.
Collapse
Affiliation(s)
- David M Francis
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Margaret P Manspeaker
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Paul A Archer
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Lauren F Sestito
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Alexander J Heiler
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Alex Schudel
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; School of Material Science and Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
13
|
Yao W, Yao J, Qian F, Que Z, Yu P, Luo T, Zheng D, Zhang Z, Tian J. Paclitaxel-loaded and folic acid-modified PLGA nanomedicine with glutathione response for the treatment of lung cancer. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1027-1036. [PMID: 34109980 DOI: 10.1093/abbs/gmab073] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Indexed: 12/19/2022] Open
Abstract
Targeted delivery and smart response of nanomedicine hold great promise for improving the therapeutic efficacy and alleviating the side effects of chemotherapy agents in cancer treatment. However, availability of only a few studies that discuss organic nanomedicines with these properties limits the development prospects of nanomedicines. In the present study, folic acid (FA)-targeted delivery and glutathione (GSH) smart responsive nanomedicine were rationally designed for paclitaxel (PTX) delivery for the treatment of lung cancer. Compared with other stimuli-responsive nanomedicines, this nanocarrier was not only sensitive to biologically relevant GSH for on-demand drug release but also biodegradable into biocompatible products after fulfilling its delivery task. The nanomedicine first entered tumor cells via FA and its receptor-mediated endocytosis. After the lysosomal escape, poly(lactic-co-glycolic acid) (PLGA) nanomedicine was triggered by a higher level of GSH and released its cargo into the tumor microenvironment. In vitro and in vivo results revealed that the PLGA nanomedicine not only inhibited the proliferation and promoted the apoptosis of lung cancer cells significantly but also possessed less toxic side effects when compared with free PTX. Therefore, the proposed drug delivery system demonstrates the potential of a multifunctional nano-platform to enhance bioavailability and reduce the side effects of chemotherapy agents.
Collapse
Affiliation(s)
- Wang Yao
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jialiang Yao
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Fangfang Qian
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Zujun Que
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Pan Yu
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Tianle Luo
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Dan Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhanxia Zhang
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jianhui Tian
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Research Center for Cancer, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| |
Collapse
|
14
|
Kadkhoda J, Akrami-Hasan-Kohal M, Tohidkia MR, Khaledi S, Davaran S, Aghanejad A. Advances in antibody nanoconjugates for diagnosis and therapy: A review of recent studies and trends. Int J Biol Macromol 2021; 185:664-678. [PMID: 34224755 DOI: 10.1016/j.ijbiomac.2021.06.191] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 01/11/2023]
Abstract
Nowadays, the targeted imaging probe and drug delivery systems are the novel breakthrough area in the nanomedicine and treatment of various diseases. Conjugation of monoclonal antibodies and their fragments on nanoparticles (NPs) have a remarkable impact on personalized medicine, such that it provides specific internalization and accumulation in the tumor microenvironment. Targeted imaging and early detection of cancer is presumably the strong participant to a diminution in mortality and recurrence of cancer disease that will be the next generation of the imaging device in clinical application. These intelligent delivery systems can deliver therapeutic agents that target cancerous tissue with minimal side effects and a wide therapeutic window. Overall, the linkage between the antibody and NPs is a critical subject and requires precise design and development. The attachment of antibody nanoconjugates (Ab-NCs) on the antigen surface shouldn't affect the function of the antibody-antigen binding. Also, the stability of the antibody nanoconjugates in blood circulation is concerned to avoid the release of drug in non-targeted regions and the possible for specific toxicity while disposal to the desired site. Here, we update the recent progress of Ab-NCs to improve early detection and cancer therapy.
Collapse
Affiliation(s)
- Jamileh Kadkhoda
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Akrami-Hasan-Kohal
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran 1591634311, Iran
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Khaledi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Mao C, Gorbet MJ, Singh A, Ranjan A, Fiering S. In situ vaccination with nanoparticles for cancer immunotherapy: understanding the immunology. Int J Hyperthermia 2021; 37:4-17. [PMID: 33455477 DOI: 10.1080/02656736.2020.1810333] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
FDA approval of anti-CTLA4 in 2011 for melanoma immunotherapy was paradigm shifting and dramatically accelerated cancer immunotherapy research. The investment and effort have been exceptionally large, with a commensurate impressive pace of discovery. Historical and current research has validated the following key points: tumors are recognized by the immune system; tumors develop an immunosuppressive environment which suppresses the antitumor immune response; successful immunotherapy must overcome that tumor-mediated immunosuppression. While cancer immunotherapy research expanded, a parallel effort developing nanoparticles (NP) for cancer diagnosis and therapy also received major investment and expanded. Initially the two efforts appeared to have minimal synergy. Systemically administered nanoparticles are rapidly ingested by phagocytic leukocytes, and therefore nanotechnologists developed strategies to avoid NP ingestion by leukocytes in order to accomplish nanoparticle accumulation in tumors rather than liver and spleen. Recently, nanotechnology and cancer immunotherapy have increasingly merged since phagocytic leukocytes are the key to reversing the local tumor immunosuppression and the tendency of NP to be phagocytosed can be exploited to manipulate phagocytes for immunotherapy. This review focuses on in situ vaccination (ISV), an immunotherapy approach that can utilize direct injection of immunostimulatory reagents, including NPs, into tumors to disrupt the local immunosuppression, stimulate effective immune response against the treated tumor, and most importantly, generate a systemic antitumor immune response to eliminate metastatic tumors. While there are many specific options for using NP for ISV (reviewed further in this special issue), this review focuses on immunology concepts needed to understand and design successful NP ISV approaches.
Collapse
Affiliation(s)
- Chenkai Mao
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Michael-Joseph Gorbet
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Akansha Singh
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Ashish Ranjan
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Norris Cotton Cancer Center, Geisel School of Medicine and Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| |
Collapse
|
16
|
Carbon dots and curcumin-loaded CD44-Targeted liposomes for imaging and tracking cancer chemotherapy: A multi-purpose tool for theranostics. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102363] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Yang S, Wallach M, Krishna A, Kurmasheva R, Sridhar S. Recent Developments in Nanomedicine for Pediatric Cancer. J Clin Med 2021; 10:1437. [PMID: 33916177 PMCID: PMC8036287 DOI: 10.3390/jcm10071437] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/20/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is the second biggest cause of death in children in the US. With the development of chemotherapy, there has been a substantial increase in the overall survival rate in the last 30 years. However, the overall mortality rate in children with cancer remains 25%, and many survivors experience a decline in overall quality of life and long-term adverse effects caused by treatments. Although cancer cells share common characteristics, pediatric cancers are different from adult cancers in their prevalence, mutation load, and drug response. Therefore, there is an urgent unmet need to develop therapeutic approaches specifically designed for children with cancer. Nanotechnology can potentially overcome the deficiencies of conventional methods of administering chemotherapy and ultimately improve clinical outcomes. The nanoparticle-based drug delivery systems can decrease the toxicity of therapy, provide a sustained or controlled drug release, improve the pharmacokinetic properties of loading contents, and achieve a targeted drug delivery with achievable modifications. Furthermore, therapeutic approaches based on combining nanoformulated drugs with novel immunotherapeutic agents are emerging. In this review, we discussed the recently developed nanotechnology-based strategies for treating blood and solid pediatric cancers.
Collapse
Affiliation(s)
- Shicheng Yang
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA;
| | - Mia Wallach
- School of Business, Northeastern University, Boston, MA 02115, USA;
| | - Apurva Krishna
- Department of Physics, Northeastern University, Boston, MA 02115, USA;
| | - Raushan Kurmasheva
- Department of Molecular Medicine, The University of Texas Health at San Antonio, San Antonio, TX 78229, USA
- Greehey Children’s Cancer Research Institute, San Antonio, TX 78229, USA
| | - Srinivas Sridhar
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA;
- Department of Physics, Northeastern University, Boston, MA 02115, USA;
- Division of Radiation Oncology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
18
|
Carter TJ, Agliardi G, Lin FY, Ellis M, Jones C, Robson M, Richard-Londt A, Southern P, Lythgoe M, Zaw Thin M, Ryzhov V, de Rosales RTM, Gruettner C, Abdollah MRA, Pedley RB, Pankhurst QA, Kalber TL, Brandner S, Quezada S, Mulholland P, Shevtsov M, Chester K. Potential of Magnetic Hyperthermia to Stimulate Localized Immune Activation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2005241. [PMID: 33734595 DOI: 10.1002/smll.202005241] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/20/2021] [Indexed: 05/27/2023]
Abstract
Magnetic hyperthermia (MH) harnesses the heat-releasing properties of superparamagnetic iron oxide nanoparticles (SPIONs) and has potential to stimulate immune activation in the tumor microenvironment whilst sparing surrounding normal tissues. To assess feasibility of localized MH in vivo, SPIONs are injected intratumorally and their fate tracked by Zirconium-89-positron emission tomography, histological analysis, and electron microscopy. Experiments show that an average of 49% (21-87%, n = 9) of SPIONs are retained within the tumor or immediately surrounding tissue. In situ heating is subsequently generated by exposure to an externally applied alternating magnetic field and monitored by thermal imaging. Tissue response to hyperthermia, measured by immunohistochemical image analysis, reveals specific and localized heat-shock protein expression following treatment. Tumor growth inhibition is also observed. To evaluate the potential effects of MH on the immune landscape, flow cytometry is used to characterize immune cells from excised tumors and draining lymph nodes. Results show an influx of activated cytotoxic T cells, alongside an increase in proliferating regulatory T cells, following treatment. Complementary changes are found in draining lymph nodes. In conclusion, results indicate that biologically reactive MH is achievable in vivo and can generate localized changes consistent with an anti-tumor immune response.
Collapse
Affiliation(s)
- Thomas J Carter
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Giulia Agliardi
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Fang-Yu Lin
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
| | - Matthew Ellis
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Cancer Sciences Unit, Cancer Research UK Centre, University of Southampton, Somers Building, Southampton, SO16 6YD, UK
| | - Clare Jones
- School of Biomedical Engineering and Imaging Sciences, King's College London (KCL), St Thomas' Hospital, London, SE1 7EH, UK
| | - Mathew Robson
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Angela Richard-Londt
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Paul Southern
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
- Resonant Circuits Limited (RCL), London, W1S 4BS, UK
| | - Mark Lythgoe
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - May Zaw Thin
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Vyacheslav Ryzhov
- NRC "Kurchatov Institute", Petersburg Nuclear Physics Institute, Gatchina, 188300, Russia
| | - Rafael T M de Rosales
- School of Biomedical Engineering and Imaging Sciences, King's College London (KCL), St Thomas' Hospital, London, SE1 7EH, UK
| | - Cordula Gruettner
- Micromod Partikeltechnologie GmbH, Friedrich-Barnewitz-Str. 4, Rostock, D-18119, Germany
| | - Maha R A Abdollah
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt (BUE), El Shorouk City, Misr- Ismalia Desert Road, 11873, Cairo, Egypt
| | - R Barbara Pedley
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Quentin A Pankhurst
- UCL Healthcare Biomagnetics Laboratory, 21 Albermarle Street, London, W1S 4BS, UK
- Resonant Circuits Limited (RCL), London, W1S 4BS, UK
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Sebastian Brandner
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Sergio Quezada
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Paul Mulholland
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Maxim Shevtsov
- NRC "Kurchatov Institute", Petersburg Nuclear Physics Institute, Gatchina, 188300, Russia
- Technical University of Munich, Klinikum Rechts der Isar, Ismaninger str. 22, Munich, 81675, Germany
| | - Kerry Chester
- UCL Cancer Institute, University College London (UCL), Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| |
Collapse
|
19
|
Chen BW, Chiu GW, He YC, Huang CY, Huang HT, Sung SY, Hsieh CL, Chang WC, Hsu MS, Wei ZH, Yao DJ. Extracellular and intracellular intermittent magnetic-fluid hyperthermia treatment of SK-Hep1 hepatocellular carcinoma cells based on magnetic nanoparticles coated with polystyrene sulfonic acid. PLoS One 2021; 16:e0245286. [PMID: 33544751 PMCID: PMC7864458 DOI: 10.1371/journal.pone.0245286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/24/2020] [Indexed: 01/27/2023] Open
Abstract
The use of magnetic nanoparticles (MNPs) magnetized on applying an alternating magnetic field (AMF) to stimulate the thermal characteristics and to induce tumor apoptosis is a currently active area of research in cancer treatment. In previous work, we developed biocompatible and superparamagnetic polystyrene-sulfonic-acid-coated magnetic nanoparticles (PSS-MNPs) as applications for magnetically labeled cell trapping, but without assessment of treatment effects on tumor diseases. In the present work, we examined PSS-MNP-induced magnetic fluid hyperthermia (MFH) on SK-Hep1 hepatocellular carcinoma (HCC) cells for lethal thermal effects with a self-made AMF system; an adjustable AMF frequency generated a variable intensity of magnetic field and induced MNP relaxation. The extracellular and intracellular MFH treatments on a SK-Hep1 cell line were implemented in vitro; the result indicates that the lethal effects were efficient and caused a significantly decreased cell viability of SK-Hep1 cells. As the PSS-MNP concentration decreased, especially in intracellular MFH treatments, the MFH effects on cells, however, largely decreased through heat spreading to the culture medium. On controlling and decreasing the volume of culture medium, the problem of heat spreading was solved. It can be consequently expected that PSS-MNPs would be a prospective agent for intracellular cancer magnetotherapy.
Collapse
Affiliation(s)
- Bo-Wei Chen
- Institute of NanoEngineering and MicroSystems, National Tsing Hua University, Hsinchu City, Taiwan
| | - Guo-Wei Chiu
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu City, Taiwan
| | - Yun-Chi He
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu City, Taiwan
| | - Chih-Yu Huang
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu City, Taiwan
| | - Hao-Ting Huang
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu City, Taiwan
| | - Shian-Ying Sung
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ling Hsieh
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chieh Chang
- Department of Neurosurgery, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Ming-Shinn Hsu
- Department of Obstetrics and Gynecology, Ching-Kuo Campus of Min-Sheng Hospital, Taoyuan, Taiwan
| | - Zung-Hang Wei
- Institute of NanoEngineering and MicroSystems, National Tsing Hua University, Hsinchu City, Taiwan
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu City, Taiwan
| | - Da-Jeng Yao
- Institute of NanoEngineering and MicroSystems, National Tsing Hua University, Hsinchu City, Taiwan
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu City, Taiwan
- * E-mail:
| |
Collapse
|
20
|
Mosaddad SA, Beigi K, Doroodizadeh T, Haghnegahdar M, Golfeshan F, Ranjbar R, Tebyanian H. Therapeutic applications of herbal/synthetic/bio-drug in oral cancer: An update. Eur J Pharmacol 2020; 890:173657. [PMID: 33096111 DOI: 10.1016/j.ejphar.2020.173657] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/01/2020] [Accepted: 10/20/2020] [Indexed: 12/18/2022]
Abstract
Oral cancer, as one of the most prevalent and invasive cancers that invade local tissue, can cause metastasis, and have high mortality. In 2018, around 355,000 worldwide oral cancers occurred and resulted in 177,000 deaths. Estimates for the year 2020 include about 53,260 new cases added to previous year's cases, and the estimated death toll from this cancer in 2020 is about 10,750 deaths more than previous years. Despite recent advances in cancer diagnosis and treatment, unfortunately, 50% of people with cancer cannot be cured. Of course, it should be remembered that the type of treatment used greatly influences patient recovery. There are not many choices when it comes to treating oral cancer. Research efforts focusing on the discovery and evolution of innovative therapeutic approaches for oral cancer are essential. Such traditional methods of treating this type of cancer like surgery and chemotherapy, have evolved dramatically during the past thirty to forty years, but they continue to cause panic among patients due to their side effects. Therefore, it is necessary to study and use drugs that are less risky for the patient as well as to provide solutions to reduce chemotherapy-induced adverse events that prevent many therapeutic risks. As mentioned above, this study examines low-risk therapies such as herbal remedies, biological drugs, and synthetic drugs in the hope that they will be useful to physicians, researchers, and scientists around the world.
Collapse
Affiliation(s)
- Seyed Ali Mosaddad
- Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kimia Beigi
- Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tayebeh Doroodizadeh
- Department of Pediatric Dentistry, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maral Haghnegahdar
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Golfeshan
- Orthodontic Research Center, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Ranjbar
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamid Tebyanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Pellas V, Hu D, Mazouzi Y, Mimoun Y, Blanchard J, Guibert C, Salmain M, Boujday S. Gold Nanorods for LSPR Biosensing: Synthesis, Coating by Silica, and Bioanalytical Applications. BIOSENSORS 2020; 10:E146. [PMID: 33080925 PMCID: PMC7603250 DOI: 10.3390/bios10100146] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022]
Abstract
Nanoparticles made of coinage metals are well known to display unique optical properties stemming from the localized surface plasmon resonance (LSPR) phenomenon, allowing their use as transducers in various biosensing configurations. While most of the reports initially dealt with spherical gold nanoparticles owing to their ease of synthesis, the interest in gold nanorods (AuNR) as plasmonic biosensors is rising steadily. These anisotropic nanoparticles exhibit, on top of the LSPR band in the blue range common with spherical nanoparticles, a longitudinal LSPR band, in all respects superior, and in particular in terms of sensitivity to the surrounding media and LSPR-biosensing. However, AuNRs synthesis and their further functionalization are less straightforward and require thorough processing. In this paper, we intend to give an up-to-date overview of gold nanorods in LSPR biosensing, starting from a critical review of the recent findings on AuNR synthesis and the main challenges related to it. We further highlight the various strategies set up to coat AuNR with a silica shell of controlled thickness and porosity compatible with LSPR-biosensing. Then, we provide a survey of the methods employed to attach various bioreceptors to AuNR. Finally, the most representative examples of AuNR-based LSPR biosensors are reviewed with a focus put on their analytical performances.
Collapse
Affiliation(s)
- Vincent Pellas
- Laboratoire de Réactivité de Surface (LRS), Sorbonne Université, CNRS, UMR 7197, 4 Place Jussieu, F-75005 Paris, France; (V.P.); (D.H.); (Y.M.); (Y.M.); (J.B.); (C.G.)
- Institut Parisien de Chimie Moléculaire (IPCM), Sorbonne Université, CNRS, 4 Place Jussieu, F-75005 Paris, France
| | - David Hu
- Laboratoire de Réactivité de Surface (LRS), Sorbonne Université, CNRS, UMR 7197, 4 Place Jussieu, F-75005 Paris, France; (V.P.); (D.H.); (Y.M.); (Y.M.); (J.B.); (C.G.)
| | - Yacine Mazouzi
- Laboratoire de Réactivité de Surface (LRS), Sorbonne Université, CNRS, UMR 7197, 4 Place Jussieu, F-75005 Paris, France; (V.P.); (D.H.); (Y.M.); (Y.M.); (J.B.); (C.G.)
| | - Yoan Mimoun
- Laboratoire de Réactivité de Surface (LRS), Sorbonne Université, CNRS, UMR 7197, 4 Place Jussieu, F-75005 Paris, France; (V.P.); (D.H.); (Y.M.); (Y.M.); (J.B.); (C.G.)
| | - Juliette Blanchard
- Laboratoire de Réactivité de Surface (LRS), Sorbonne Université, CNRS, UMR 7197, 4 Place Jussieu, F-75005 Paris, France; (V.P.); (D.H.); (Y.M.); (Y.M.); (J.B.); (C.G.)
| | - Clément Guibert
- Laboratoire de Réactivité de Surface (LRS), Sorbonne Université, CNRS, UMR 7197, 4 Place Jussieu, F-75005 Paris, France; (V.P.); (D.H.); (Y.M.); (Y.M.); (J.B.); (C.G.)
| | - Michèle Salmain
- Institut Parisien de Chimie Moléculaire (IPCM), Sorbonne Université, CNRS, 4 Place Jussieu, F-75005 Paris, France
| | - Souhir Boujday
- Laboratoire de Réactivité de Surface (LRS), Sorbonne Université, CNRS, UMR 7197, 4 Place Jussieu, F-75005 Paris, France; (V.P.); (D.H.); (Y.M.); (Y.M.); (J.B.); (C.G.)
| |
Collapse
|
22
|
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]
|
23
|
Bhattacharya DS, Svechkarev D, Bapat A, Patil P, Hollingsworth MA, Mohs AM. Sulfation modulates the targeting properties of hyaluronic acid to P-selectin and CD44. ACS Biomater Sci Eng 2020; 6:3585-3598. [PMID: 32617404 PMCID: PMC7331950 DOI: 10.1021/acsbiomaterials.0c00115] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Many targeting strategies can be employed to direct nanoparticles to tumors for imaging and therapy. However, tumors display a dynamic, heterogeneous microenvironment that undergoes spatiotemporal changes, including the expression of targetable cell-surface biomarkers. Here, we develop a nanoparticle system to effectively target two receptors overexpressed in the microenvironment of aggressive tumors. Hyaluronic acid (HA) was regioselectivity modified using a multi-step synthetic approach to alter binding specificities for CD44 and P-selectin to tumor cell interaction. The dual-targeting strategy utilizes sulfate modifications on HA that targets P-selectin, in addition to native targeting of CD44, which exploits spatiotemporal alterations in the expression patterns of these two receptors in cancer sites. Using biophysical characterization and in vitro studies, we demonstrate that modified HA nanoparticles effectively targets both P-selectin+ and CD44+ cells, which lays the groundwork for future in vivo biomedical applications.
Collapse
Affiliation(s)
- Deep S. Bhattacharya
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Denis Svechkarev
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Aishwarya Bapat
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Prathamesh Patil
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Aaron M. Mohs
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
24
|
Park J, Chariou PL, Steinmetz NF. Site-Specific Antibody Conjugation Strategy to Functionalize Virus-Based Nanoparticles. Bioconjug Chem 2020; 31:1408-1416. [PMID: 32281790 PMCID: PMC8085887 DOI: 10.1021/acs.bioconjchem.0c00118] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Amine/thiol-reactive chemistries are commonly used to conjugate antibodies to pharmaceuticals or nanoparticles. Yet, these conjugation strategies often result in unfavorable outcomes such as heterogeneous antibody display with hindered biological activity or aggregation due to multivalent interactions of the antibody and nanoparticles. Here, we report the application of a site-specific and enzymatically driven antibody conjugation strategy to functionalize virus-based nanoparticles (VNPs). Specifically, an azide-handle was introduced into the Fc region of a set of immunoglobulins using a two-step enzymatic reaction: (1) cleavage of N-linked glycan in the Fc region by a glycosidase and (2) conjugation of a chemically reactive linker (containing an azide functional handle) using a microbial transglutaminase. Conjugation of the azide-functional antibodies to several VNPs was achieved by making use of strain-promoted azide-alkyne cycloaddition. We report the conjugation of three immunoglobulin (IgG) isotypes (human IgG from sera, anti-CD47 Rat IgG2a, κ, and Trastuzumab recombinant humanized IgG1, κ) to the plant virus cowpea mosaic virus (CPMV) and the lysine mutant of tobacco mosaic virus (TMVlys) as well as bacteriophage Qβ. Site-specific conjugation resulted in stable and functional antibody-VNP conjugates. In stark contrast, the use of heterobifunctional linkers targeting thiols and amines on the antibodies and VNPs, respectively, led to aggregation due to nonspecific and multivalent coupling between the antibodies and VNPs. We demonstrate that antibody-VNP conjugates were functional, and Trastuzumab-displaying VNPs targeted HER2-positive SKOV-3 human ovarian cancer cells. This bioconjugation strategy adds to the portfolio of methods that can be used for designing functional antibody-VNP conjugates.
Collapse
|
25
|
Chariou PL, Ortega-Rivera OA, Steinmetz NF. Nanocarriers for the Delivery of Medical, Veterinary, and Agricultural Active Ingredients. ACS NANO 2020; 14:2678-2701. [PMID: 32125825 PMCID: PMC8085836 DOI: 10.1021/acsnano.0c00173] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Nanocarrier-based delivery systems can be used to increase the safety and efficacy of active ingredients in medical, veterinary, or agricultural applications, particularly when such ingredients are unstable, sparingly soluble, or cause off-target effects. In this review, we highlight the diversity of nanocarrier materials and their key advantages compared to free active ingredients. We discuss current trends based on peer-reviewed research articles, patent applications, clinical trials, and the nanocarrier formulations already approved by regulatory bodies. Although most nanocarriers have been engineered to combat cancer, the number of formulations developed for other purposes is growing rapidly, especially those for the treatment of infectious diseases and parasites affecting humans, livestock, and companion animals. The regulation and prohibition of many pesticides have also fueled research to develop targeted pesticide delivery systems based on nanocarriers, which maximize efficacy while minimizing the environmental impact of agrochemicals.
Collapse
|
26
|
Essa D, Kondiah PPD, Choonara YE, Pillay V. The Design of Poly(lactide-co-glycolide) Nanocarriers for Medical Applications. Front Bioeng Biotechnol 2020; 8:48. [PMID: 32117928 PMCID: PMC7026499 DOI: 10.3389/fbioe.2020.00048] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/22/2020] [Indexed: 12/19/2022] Open
Abstract
Polymeric biomaterials have found widespread applications in nanomedicine, and poly(lactide-co-glycolide), (PLGA) in particular has been successfully implemented in numerous drug delivery formulations due to its synthetic malleability and biocompatibility. However, the need for preconception in these formulations is increasing, and this can be achieved by selection and elimination of design variables in order for these systems to be tailored for their specific applications. The starting materials and preparation methods have been shown to influence various parameters of PLGA-based nanocarriers and their implementation in drug delivery systems, while the implementation of computational simulations as a component of formulation studies can provide valuable information on their characteristics. This review provides a critical summary of the synthesis and applications of PLGA-based systems in bio-medicine and outlines experimental and computational design considerations of these systems.
Collapse
Affiliation(s)
| | | | | | - Viness Pillay
- Wits Advanced Drug Delivery Platform, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
27
|
Pant K, Neuber C, Zarschler K, Wodtke J, Meister S, Haag R, Pietzsch J, Stephan H. Active Targeting of Dendritic Polyglycerols for Diagnostic Cancer Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1905013. [PMID: 31880080 DOI: 10.1002/smll.201905013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/15/2019] [Indexed: 06/10/2023]
Abstract
Active tumor targeting involves the decoration of nanomaterials (NMs) with oncotropic vector biomolecules that selectively recognize certain antigens on malignant cells or in the tumor microenvironment. This strategy can facilitate intracellular uptake of NM through specific interactions such as receptor-mediated endocytosis and can lead to prolonged retention in the malignant tissues by preventing rapid efflux from the tumor. Here, the design of actively targeting, renally excretible bimodal dendritic polyglycerols (dPGs) for diagnostic cancer imaging is described. Single-domain antibodies (sdAbs) specifically binding to the epidermal growth factor receptor (EGFR) are employed herein as targeting warheads owing to their small size and high affinity for their corresponding antigen. The dPGs equipped with EGFR-targeting feature are compared head-to-head with their nontargeting counterparts in terms of interaction with EGFR-overexpressing cells in vitro as well as accumulation at receptor-positive tumors in vivo. Experimental results reveal a higher specificity and preferential tumor accumulation for the α-EGFR dPGs, resulting from the introduction of active targeting capabilities on their backbone. These results highlight the potential for improving the tumor uptake properties of dPGs by strategic use of sdAb functionalization, which can ultimately prove useful to the development of ultrasmall NM with highly specific tumor accumulation.
Collapse
Affiliation(s)
- Kritee Pant
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden - Rossendorf, Bautzner Landstrasse 400, D-01328, Dresden, Germany
| | - Christin Neuber
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden - Rossendorf, Bautzner Landstrasse 400, D-01328, Dresden, Germany
| | - Kristof Zarschler
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden - Rossendorf, Bautzner Landstrasse 400, D-01328, Dresden, Germany
| | - Johanna Wodtke
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden - Rossendorf, Bautzner Landstrasse 400, D-01328, Dresden, Germany
| | - Sebastian Meister
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden - Rossendorf, Bautzner Landstrasse 400, D-01328, Dresden, Germany
| | - Rainer Haag
- Organische Chemie, Institut für Chemie und Biochemie, Freie Universität Berlin, Takustrasse 3, D-14195, Berlin, Germany
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden - Rossendorf, Bautzner Landstrasse 400, D-01328, Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, D-01062, Dresden, Germany
| | - Holger Stephan
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden - Rossendorf, Bautzner Landstrasse 400, D-01328, Dresden, Germany
| |
Collapse
|
28
|
Marques AC, Costa PJ, Velho S, Amaral MH. Functionalizing nanoparticles with cancer-targeting antibodies: A comparison of strategies. J Control Release 2020; 320:180-200. [PMID: 31978444 DOI: 10.1016/j.jconrel.2020.01.035] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 01/07/2023]
Abstract
Standard cancer therapies sometimes fail to deliver chemotherapeutic drugs to tumor cells in a safe and effective manner. Nanotechnology takes the lead in providing new therapeutic options for cancer due to major potential for selective targeting and controlled drug release. Antibodies and antibody fragments are attracting much attention as a source of targeting ligands to bind specific receptors that are overexpressed on cancer cells. Therefore, researchers are devoting time and effort to develop targeting strategies based on nanoparticles functionalized with antibodies, which hold great promise to enhance therapeutic efficacy and circumvent severe side effects. Several methods have been described to immobilize antibodies on the surface of nanoparticles. However, selecting the most appropriate for each application is challenging but also imperative to preserve antigen binding ability and yield stable antibody-conjugated nanoparticles. From this perspective, we aim to provide considerable knowledge on the most widely used methods of functionalization that can be helpful for decision-making and design of conjugation protocols as well. This review summarizes adsorption, covalent conjugation (carbodiimide, maleimide and "click" chemistries) and biotin-avidin interaction, while discussing the advantages, limitations and relevant therapeutic approaches currently under investigation.
Collapse
Affiliation(s)
- A C Marques
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| | - P J Costa
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - S Velho
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, R. Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
| | - M H Amaral
- UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto (FFUP), R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
29
|
Ngo-Duc TT, Alibay Z, Plank JM, Cheeney JE, Haberer ED. Gold-Decorated M13 I-Forms and S-Forms for Targeted Photothermal Lysis of Bacteria. ACS APPLIED MATERIALS & INTERFACES 2020; 12:126-134. [PMID: 31800209 DOI: 10.1021/acsami.9b15682] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
With the emergence of multidrug-resistant bacteria, photothermal therapy has been proposed as an alternative to antibiotics for targeting and killing pathogens. In this study, two M13 bacteriophage polymorphs were studied as nanoscaffolds for plasmonic bactericidal agents. Receptor-binding proteins found on the pIII minor coat protein targeted Escherichia coli bacteria with F-pili (F+ strain), while a gold-binding peptide motif displayed on the pVIII major coat protein templated Au nanoparticles. Temperature-dependent exposure to a chloroform-water interface transformed the native filamentous phage into either rod-like or spheroid structures. The morphology, geometry, and size of the polymorphs, as well as the receptor-binding protein and host cell receptor interaction were studied using electron microscopy. Au/template structures were formed through incubation with Au colloid, and optical absorbance was measured. Despite the closely packed Au nanoparticle layer on the surface the viral scaffolds, electron microscopy confirmed that host receptor affinity was retained. Photothermal bactericidal studies were performed using 532 nm laser irradiation with a variety of powers and exposure times. Bacterial viability was assessed using colony count. With the shape-modified M13 scaffolds, up to 64% of E. coli were killed within 20 min. These studies demonstrate the promise of i-form and s-form polymorphs for the directed plasmonic-based photothermal killing of bacteria.
Collapse
Affiliation(s)
- Tam-Triet Ngo-Duc
- Materials Science and Engineering Program , University of California , Riverside 92521 , United States
| | - Zaira Alibay
- Materials Science and Engineering Program , University of California , Riverside 92521 , United States
| | - Joshua M Plank
- Department of Electrical and Computer Engineering , University of California , Riverside 92521 , United States
| | - Joseph Earl Cheeney
- Materials Science and Engineering Program , University of California , Riverside 92521 , United States
| | - Elaine D Haberer
- Materials Science and Engineering Program , University of California , Riverside 92521 , United States
- Department of Electrical and Computer Engineering , University of California , Riverside 92521 , United States
| |
Collapse
|
30
|
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
|
31
|
Mendes LP, Rostamizadeh K, Gollomp K, Myerson JW, Marcos-Contreras OA, Zamora M, Luther E, Brenner JS, Filipczak N, Li X, Torchilin VP. Monoclonal antibody 2C5 specifically targets neutrophil extracellular traps. MAbs 2020; 12:1850394. [PMID: 33323006 PMCID: PMC7755171 DOI: 10.1080/19420862.2020.1850394] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 11/03/2020] [Accepted: 11/10/2020] [Indexed: 10/27/2022] Open
Abstract
Neutrophils can release DNA and granular cytoplasmic proteins that form smooth filaments of stacked nucleosomes (NS). These structures, called neutrophil extracellular traps (NETs), are involved in multiple pathological processes, and NET formation and removal are clinically significant. The monoclonal antibody 2C5 has strong specificity toward intact NS but not to individual NS components, indicating that 2C5 could potentially target NS in NETs. In this study, NETs were generated in vitro using neutrophils and HL-60 cells differentiated into granulocyte-like cells. The specificity of 2C5 toward NETs was evaluated by ELISA, which showed that it binds to NETs with the specificity similar to that for purified nucleohistone substrate. Immunofluorescence showed that 2C5 stains NETs in both static and perfused microfluidic cell cultures, even after NET compaction. Modification of liposomes with 2C5 dramatically enhanced liposome association with NETs. Our results suggest that 2C5 could be used to identify and visualize NETs and serve as a ligand for NET-targeted diagnostics and therapies.
Collapse
Affiliation(s)
- Livia P. Mendes
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
| | - Kobra Rostamizadeh
- Zanjan Pharmaceutical Nanotechnology Research Center, School of Pharmacy, Pharmaceutical Biomaterials Department, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kandace Gollomp
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jacob W. Myerson
- Department of Systems Pharmacology and Translational Therapeutics, University of Philadelphia, Philadelphia, PA, USA
| | - Oscar A. Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, University of Philadelphia, Philadelphia, PA, USA
| | - Marco Zamora
- Department of Systems Pharmacology and Translational Therapeutics, University of Philadelphia, Philadelphia, PA, USA
| | - Ed Luther
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Jacob S. Brenner
- Department of Systems Pharmacology and Translational Therapeutics, University of Philadelphia, Philadelphia, PA, USA
- Pulmonary, Allergy, & Critical Care Division, University of Philadelphia, PA, USA
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
| | - Xiang Li
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Vladimir P. Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
- Department of Oncology, Radiotherapy and Plastic Surgery, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
32
|
Huang X, Lin H, Huang F, Xie Y, Wong KH, Chen X, Wu D, Lu A, Yang Z. Targeting Approaches of Nanomedicines in Acute Myeloid Leukemia. Dose Response 2019; 17:1559325819887048. [PMID: 31853234 PMCID: PMC6906351 DOI: 10.1177/1559325819887048] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/10/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy, which is commonly
associated with high incidence and mortality among adult patients. The standard
induction regimen for AML has been substantially unchanged over the past 40
years, for which novel nanomedicines have represented a promising strategy in
AML therapies. Despite developments of multiple nanoparticles formulated with
drugs or genes, less there is not much information available about approaches in
AML is available. This review presents an overview of nanomedicines currently
being evaluated in AML. First, it briefly summarized conventional chemotherapies
in use. Second, nanomedicines presently ongoing in clinical trials or
preclinical researches were classified and described, with illustrative examples
from recent literatures. Finally, limitations and potential safety issues
concerns in clinical translation of AML treatment were discussed as well.
Collapse
Affiliation(s)
- Xiao Huang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hai Lin
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Feng Huang
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuning Xie
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ka Hong Wong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xiaoyu Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Dongyue Wu
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
33
|
Shreffler JW, Pullan JE, Dailey KM, Mallik S, Brooks AE. Overcoming Hurdles in Nanoparticle Clinical Translation: The Influence of Experimental Design and Surface Modification. Int J Mol Sci 2019; 20:E6056. [PMID: 31801303 PMCID: PMC6928924 DOI: 10.3390/ijms20236056] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/23/2019] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles are becoming an increasingly popular tool for biomedical imaging and drug delivery. While the prevalence of nanoparticle drug-delivery systems reported in the literature increases yearly, relatively little translation from the bench to the bedside has occurred. It is crucial for the scientific community to recognize this shortcoming and re-evaluate standard practices in the field, to increase clinical translatability. Currently, nanoparticle drug-delivery systems are designed to increase circulation, target disease states, enhance retention in diseased tissues, and provide targeted payload release. To manage these demands, the surface of the particle is often modified with a variety of chemical and biological moieties, including PEG, tumor targeting peptides, and environmentally responsive linkers. Regardless of the surface modifications, the nano-bio interface, which is mediated by opsonization and the protein corona, often remains problematic. While fabrication and assessment techniques for nanoparticles have seen continued advances, a thorough evaluation of the particle's interaction with the immune system has lagged behind, seemingly taking a backseat to particle characterization. This review explores current limitations in the evaluation of surface-modified nanoparticle biocompatibility and in vivo model selection, suggesting a promising standardized pathway to clinical translation.
Collapse
Affiliation(s)
| | | | | | | | - Amanda E. Brooks
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA; (J.W.S.); (J.E.P.); (K.M.D.); (S.M.)
| |
Collapse
|
34
|
Shilovskiy IP, Dyneva ME, Kurbacheva OM, Kudlay DA, Khaitov MR. The Role of Interleukin-37 in the Pathogenesis of Allergic Diseases. Acta Naturae 2019; 11:54-64. [PMID: 31993235 PMCID: PMC6977961 DOI: 10.32607/20758251-2019-11-4-54-64] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/25/2019] [Indexed: 12/17/2022] Open
Abstract
Cytokines of the interleukin-1 (IL-1) family play an important role in the realization of the protective functions of innate immunity and are the key mediators involved in the pathogenesis of a wide range of diseases, including various manifestations of allergy. The IL-1 family includes more than 11 members. However, the functions of many of them remain to be elucidated. Recently, new members of the IL-1 family have been discovered. In 2000, several independent research groups reported the discovery of a new interleukin of this family, which was named IL-37, or IL-1F7 (according to the new nomenclature). IL-37 was assigned to the IL-1 family based on its structural similarity with other members of this family. The study of its biological properties showed that its activity changes in inflammatory diseases, such as rheumatoid arthritis, psoriasis, as well as allergic diseases (allergic rhinitis, bronchial asthma, and atopic dermatitis). However, unlike most members of the IL-1 family, IL-37 acts as a negative regulator of inflammation. Activation of IL-37 suppresses inflammation, resulting in the suppression of inflammatory cytokines and chemokines, which in turn prevents infiltration of pro-inflammatory cells, mainly eosinophils and neutrophils. The exact molecular and cellular mechanisms of the anti-inflammatory effect of IL-37 in the development of allergic diseases (AD) have not been fully studied. This review summarizes and analyzes the accumulated experimental data on the role of IL-37 in the pathogenesis of AD, such as allergic rhinitis, bronchial asthma, and atopic dermatitis.
Collapse
Affiliation(s)
- I. P. Shilovskiy
- National Research Center – Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, 115522 Russia
| | - M. E. Dyneva
- National Research Center – Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, 115522 Russia
| | - O. M. Kurbacheva
- National Research Center – Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, 115522 Russia
| | - D. A. Kudlay
- National Research Center – Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, 115522 Russia
| | - M. R. Khaitov
- National Research Center – Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, 115522 Russia
| |
Collapse
|
35
|
Shilova ON, Deyev SM. DARPins: Promising Scaffolds for Theranostics. Acta Naturae 2019; 11:42-53. [PMID: 31993234 PMCID: PMC6977956 DOI: 10.32607/20758251-2019-11-4-42-53] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 10/08/2019] [Indexed: 12/22/2022] Open
Abstract
Monoclonal antibodies are the classical basis for targeted therapy, but the development of alternative binding proteins has made it possible to use non-immunoglobulin proteins as targeting modules. The advantages of DARPins, scaffold proteins based on ankyrin repeats, over antibodies are as follows: small size, stability over a wide range of temperatures and pH values, low aggregation tendency, and ease of production in heterologous expression systems. The differences in the structure of the paratope of DARPin and antibodies broaden the spectrum of target molecules, while the ease of creating hybrid fusion proteins allows one to obtain bispecific and multivalent constructs. In this article, we summarize recent data on the development of therapeutic and imaging compounds based on DARPins.
Collapse
Affiliation(s)
- O. N. Shilova
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow
| | - S. M. Deyev
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow
| |
Collapse
|
36
|
Pearce AK, O'Reilly RK. Insights into Active Targeting of Nanoparticles in Drug Delivery: Advances in Clinical Studies and Design Considerations for Cancer Nanomedicine. Bioconjug Chem 2019; 30:2300-2311. [PMID: 31441642 DOI: 10.1021/acs.bioconjchem.9b00456] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanomedicine is a promising strategy for improving clinical outcomes for cancer therapies, by improving drug efficacy through enhanced delivery to disease sites. It is of importance for ultimate clinical success to consider the contributing factors to achieving this goal, such as size, chemistry, and functionality of nanoparticle delivery systems, and how these parameters influence tumor localization and uptake. This Topical Review will first discuss the evolution and progress of nanoparticles for cancer drug delivery and the current challenges that remain to be addressed. Strategies for overcoming the limitations of passive targeting through active targeting approaches, and the current state of such nanomedicines in the clinic will be highlighted. Finally, novel approaches toward the design of active targeted nanoparticles building on our growing understanding of nanobio interactions are considered, in order to shed light on future design considerations for accelerating clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Amanda K Pearce
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham B15 2TT , United Kingdom
| | - Rachel K O'Reilly
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham B15 2TT , United Kingdom
| |
Collapse
|
37
|
Sousa AR, Oliveira MJ, Sarmento B. Impact of CEA-targeting Nanoparticles for Drug Delivery in Colorectal Cancer. J Pharmacol Exp Ther 2019; 370:657-670. [PMID: 30670480 DOI: 10.1124/jpet.118.254441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/18/2019] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common causes of cancer-related death in the world, mainly owing to distant metastasis events. Developing targeted strategies to treat and follow individuals in more developed stages is needed. The carcinoembryonic antigen (CEA) is a cell surface-overexpressed glycoprotein in most CRC patients, and the evaluation of its serum levels is recommended in the clinic. These reasons motivated the production of CEA-targeted nanotechnologies for monitorization of CRC progression, but only a few centers have reported their use for drug delivery. The cellular internalization of CEA-linked nanosystems occurs by the natural recycling of the CEA itself, enabling longer retention and sustained release of the cargo. The functionalization of nanoparticles with lower affinity ligands for CEA is possibly the best choice to avoid their binding to the soluble CEA. Here, we also highlight the use of nanoparticles made of poly(lactic-co-glycolic acid) (PLGA) polymer, a well known material, owing to its biocompatibility and low toxicity. This work offers support to the contribution of antibody fragment-functionalized nanoparticles as promising high affinity molecules to decorate nanosystems. The linkers and conjugation chemistries chosen for ligand-nanoparticle coupling will be addressed herein as an elements essential to the modulation of nanosystem features. This review, to our knowledge, is the first that focuses on CEA-targeted nanotechnologies to serve colorectal cancer therapy and monitorization.
Collapse
Affiliation(s)
- Ana Rita Sousa
- Instituto de Investigação e Inovação em Saúde (A.R.S., M.J.O., B.S.), Instituto de Engenharia Biomédica (A.R.S., M.J.O., B.S.), Instituto de Ciências Biomédicas Abel Salazar (A.R.S., M.J.O.), and Faculdade de Medicina da (M.J.O.), Universidade do Porto, Porto, Portugal; Instituto Português de Oncologia do Porto, Porto, Portugal (A.R.S.); and Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde & Instituto Universitário de Ciências da Saúde, Gandra, Portugal (B.S.)
| | - Maria José Oliveira
- Instituto de Investigação e Inovação em Saúde (A.R.S., M.J.O., B.S.), Instituto de Engenharia Biomédica (A.R.S., M.J.O., B.S.), Instituto de Ciências Biomédicas Abel Salazar (A.R.S., M.J.O.), and Faculdade de Medicina da (M.J.O.), Universidade do Porto, Porto, Portugal; Instituto Português de Oncologia do Porto, Porto, Portugal (A.R.S.); and Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde & Instituto Universitário de Ciências da Saúde, Gandra, Portugal (B.S.)
| | - Bruno Sarmento
- Instituto de Investigação e Inovação em Saúde (A.R.S., M.J.O., B.S.), Instituto de Engenharia Biomédica (A.R.S., M.J.O., B.S.), Instituto de Ciências Biomédicas Abel Salazar (A.R.S., M.J.O.), and Faculdade de Medicina da (M.J.O.), Universidade do Porto, Porto, Portugal; Instituto Português de Oncologia do Porto, Porto, Portugal (A.R.S.); and Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde & Instituto Universitário de Ciências da Saúde, Gandra, Portugal (B.S.)
| |
Collapse
|
38
|
Gong BS, Wang R, Xu HX, Miao MY, Yao ZZ. Nanotherapy Targeting the Tumor Microenvironment. Curr Cancer Drug Targets 2019; 19:525-533. [DOI: 10.2174/1568009619666181220103714] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/11/2018] [Accepted: 11/13/2018] [Indexed: 01/24/2023]
Abstract
Cancer is characterized by high mortality and low curability. Recent studies have shown
that the mechanism of tumor resistance involves not only endogenous changes to tumor cells, but
also to the tumor microenvironment (TME), which provides the necessary conditions for the growth,
invasion, and metastasis of cancer cells, akin to Stephen Paget’s hypothesis of “seed and soil.”
Hence, the TME is a significant target for cancer therapy via nanoparticles, which can carry different
kinds of drugs targeting different types or stages of tumors. The key step of nanotherapy is the
achievement of accurate active or passive targeting to trigger drugs precisely at tumor cells, with less
toxicity and fewer side effects. With deepened understanding of the tumor microenvironment and
rapid development of the nanomaterial industry, the mechanisms of nanotherapy could be individualized
according to the specific TME characteristics, including low pH, cancer-associated fibroblasts
(CAFs), and increased expression of metalloproteinase. However, some abnormal features of the
TME limit drugs from reaching all tumor cells in lethal concentrations, and the characteristics of tumors
vary in numerous ways, resulting in great challenges for the clinical application of nanotherapy.
In this review, we discuss the essential role of the tumor microenvironment in the genesis and
development of tumors, as well as the measures required to improve the therapeutic effects of tumor
microenvironment-targeting nanoparticles and ways to reduce damage to normal tissue.
Collapse
Affiliation(s)
- Bo-Shen Gong
- Administration Office for Undergraduates, Second Military Medical University, Shanghai, 200433, China
| | - Rui Wang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai, 200433, China
| | - Hong-Xia Xu
- Department of Nutrition, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, 400042, China
| | - Ming-Yong Miao
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai, 200433, China
| | - Zhen-Zhen Yao
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|
39
|
Anderson SD, Gwenin VV, Gwenin CD. Magnetic Functionalized Nanoparticles for Biomedical, Drug Delivery and Imaging Applications. NANOSCALE RESEARCH LETTERS 2019; 14:188. [PMID: 31147786 PMCID: PMC6542970 DOI: 10.1186/s11671-019-3019-6] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/17/2019] [Indexed: 05/12/2023]
Abstract
Medicine is constantly looking for new and improved treatments for diseases, which need to have a high efficacy and be cost-effective, creating a large demand on scientific research to discover such new treatments. One important aspect of any treatment is the ability to be able to target only the illness and not cause harm to another healthy part of the body. For this reason, metallic nanoparticles have been and are currently being extensively researched for their possible medical uses, including medical imaging, antibacterial and antiviral applications. Superparamagnetic metal nanoparticles possess properties that allow them to be directed around the body with a magnetic field or directed to a magnetic implant, which opens up the potential to conjugate various bio-cargos to the nanoparticles that could then be directed for treatment in the body. Here we report on some of the current bio-medical applications of various metal nanoparticles, including single metal nanoparticles, functionalized metal nanoparticles, and core-shell metal nanoparticles using a core of Fe3O4 as well as synthesis methods of these core-shell nanoparticles.
Collapse
Affiliation(s)
- Simon D Anderson
- School of Natural Sciences, College of Environmental Sciences and Engineering, Bangor University, Bangor, LL57 2UW, UK
| | - Vanessa V Gwenin
- School of Natural Sciences, College of Environmental Sciences and Engineering, Bangor University, Bangor, LL57 2UW, UK
| | - Christopher D Gwenin
- School of Natural Sciences, College of Environmental Sciences and Engineering, Bangor University, Bangor, LL57 2UW, UK.
| |
Collapse
|
40
|
Cheng CT, Castro G, Liu CH, Lau P. Advanced nanotechnology: An arsenal to enhance immunotherapy in fighting cancer. Clin Chim Acta 2019; 492:12-19. [PMID: 30711524 DOI: 10.1016/j.cca.2019.01.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/30/2019] [Accepted: 01/30/2019] [Indexed: 12/15/2022]
Abstract
Cancer remains a major disease process with considerable healthcare and socioeconomic impact worldwide. Unfortunately, standard treatments using chemotherapy often do not effectively control cancer progression or prevent relapse. Over the past decades, the development of targeted therapies has substantially improved outcomes. Recently, immunotherapy has emerged as a new alternative for more effective cancer treatment and may even bring hope of a cure. Cancer immunotherapy functions by reinforcing a patient's immune defense system to fight the disease. Clinically, promising immunotherapy approaches have, however, been limited by unpredictable response and strong adverse effects. A drug delivery system (DDS) that effectively targets tumor and reduces drug exposure to normal tissue would mitigate these limitations. In this regard, nanotechnology has been intensively studied as a DDS for targeting tumors with various oncologic drugs. Several have resulted in improved treatment and outcome. Research has shown that nanoparticle drug delivery technologies can also be applied to immunotherapy. In this review, the current state of nanotechnology will be discussed. Because most cancer immunotherapies approved in recent years are protein drugs, this article will focus on a micellar nanocomplex (MNC) technology, a DDS platform especially suited for targeted delivery of these therapeutics to solid tumors.
Collapse
Affiliation(s)
- Chun-Ting Cheng
- Suntec Medical, Inc., 28F., No. 27-2, Sec. 2, Zhongzheng E. Rd., Tamsui Dist., New Taipei City 251, Taiwan
| | - Gabriel Castro
- Suntec Medical, Inc., 28F., No. 27-2, Sec. 2, Zhongzheng E. Rd., Tamsui Dist., New Taipei City 251, Taiwan
| | - Chun-Hsin Liu
- Suntec Medical, Inc., 28F., No. 27-2, Sec. 2, Zhongzheng E. Rd., Tamsui Dist., New Taipei City 251, Taiwan
| | - Pauline Lau
- Suntec Medical, Inc., 28F., No. 27-2, Sec. 2, Zhongzheng E. Rd., Tamsui Dist., New Taipei City 251, Taiwan; Suntec Medical, Inc, 4008 Blair Ridge Drive, Chino Hills, CA 91709, USA.
| |
Collapse
|
41
|
Nifontova G, Ramos-Gomes F, Baryshnikova M, Alves F, Nabiev I, Sukhanova A. Cancer Cell Targeting With Functionalized Quantum Dot-Encoded Polyelectrolyte Microcapsules. Front Chem 2019; 7:34. [PMID: 30761294 PMCID: PMC6363708 DOI: 10.3389/fchem.2019.00034] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/14/2019] [Indexed: 01/13/2023] Open
Abstract
Imaging agents and drug carriers are commonly targeted toward cancer cell through functionalization with specific recognition molecules. Quantum dots (QDs) are fluorescent semiconductor nanocrystals whose extraordinary brightness and photostability make them attractive for direct fluorescent labeling of biomolecules or optical encoding of the membranes and cells. Here, we analyse the cytotoxicity of QD-encoded microcapsules, validate an approach to the activation of the microcapsule's surface for further functionalization with monoclonal antibody Trastuzumab, a humanized monoclonal antibody targeting the extracellular domain of the human epidermal growth factor receptor 2 (HER2) and already in clinical use for the treatment of HER2 positive breast cancer. In addition, we characterize the cell-specific targeting activity of the resultant bio-conjugate by immunofluorescence assay (IFA) and real-time analysis of interaction of the conjugates with live HER2 overexpressing human breast cancer cells. We demonstrate, that encapsulation of QDs into the polymer shell using the layer-by-layer deposition method yields highly fluorescent polyelectrolyte microcapsules with a homogeneous size distribution and biocompatibility upon in vitro treatment of cancer cells. Carbodiimide surface activation ensures optimal disperse and optical characteristics of the QD-encoded microcapsules before antibody conjugation. The prepared conjugates of the microcapsules with cancer-specific monoclonal antibody targeting HER2 provide sufficiently sensitive and specific antibody-mediated binding of the microcapsules with live cancer cells, which demonstrated their potential as prospective cancer cell–targeting agents.
Collapse
Affiliation(s)
- Galina Nifontova
- Laboratory of Nano-Bioengineering, Moscow Engineering Physics Institute, National Research Nuclear University MEPhI, Moscow, Russia
| | - Fernanda Ramos-Gomes
- Translational Molecular Imaging, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany
| | - Maria Baryshnikova
- Laboratory of Nano-Bioengineering, Moscow Engineering Physics Institute, National Research Nuclear University MEPhI, Moscow, Russia.,N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Institute of Experimental Diagnostic and Biotherapy, Moscow, Russia
| | - Frauke Alves
- Translational Molecular Imaging, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany.,Clinic of Haematology and Medical Oncology, Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Igor Nabiev
- Laboratory of Nano-Bioengineering, Moscow Engineering Physics Institute, National Research Nuclear University MEPhI, Moscow, Russia.,Laboratoire de Recherche en Nanosciences (LRN-EA4682), Université de Reims Champagne-Ardenne, Reims, France
| | - Alyona Sukhanova
- Laboratory of Nano-Bioengineering, Moscow Engineering Physics Institute, National Research Nuclear University MEPhI, Moscow, Russia.,Laboratoire de Recherche en Nanosciences (LRN-EA4682), Université de Reims Champagne-Ardenne, Reims, France
| |
Collapse
|
42
|
Makhlouf A, Hajdu I, Hartimath SV, Alizadeh E, Wharton K, Wasan KM, Badea I, Fonge H. 111In-Labeled Glycoprotein Nonmetastatic b (GPNMB) Targeted Gemini Surfactant-Based Nanoparticles against Melanoma: In Vitro Characterization and in Vivo Evaluation in Melanoma Mouse Xenograft Model. Mol Pharm 2019; 16:542-551. [DOI: 10.1021/acs.molpharmaceut.8b00831] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Amal Makhlouf
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini, 12411 Cairo, Egypt
| | - Istvan Hajdu
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Siddesh V. Hartimath
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, 103 University Drive, Saskatoon, Saskatchewan S7N 0W8, Canada
- Saskatchewan Centre for Cyclotron Sciences (SCCS), The Fedoruk Centre, 120 Maintenance Road, Saskatoon, Saskatchewan S7N 5C4, Canada
| | - Elahe Alizadeh
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, 103 University Drive, Saskatoon, Saskatchewan S7N 0W8, Canada
- Saskatchewan Centre for Cyclotron Sciences (SCCS), The Fedoruk Centre, 120 Maintenance Road, Saskatoon, Saskatchewan S7N 5C4, Canada
| | - Kayla Wharton
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Kishor M. Wasan
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Ildiko Badea
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Humphrey Fonge
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, 103 University Drive, Saskatoon, Saskatchewan S7N 0W8, Canada
- Saskatchewan Centre for Cyclotron Sciences (SCCS), The Fedoruk Centre, 120 Maintenance Road, Saskatoon, Saskatchewan S7N 5C4, Canada
- Department of Medical Imaging, Royal University Hospital Saskatoon, 103 University Drive, Saskatoon, Saskatchewan S7N 0W8, Canada
| |
Collapse
|
43
|
Horky P, Skalickova S, Baholet D, Skladanka J. Nanoparticles as a Solution for Eliminating the Risk of Mycotoxins. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E727. [PMID: 30223519 PMCID: PMC6164963 DOI: 10.3390/nano8090727] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/04/2018] [Accepted: 09/04/2018] [Indexed: 12/21/2022]
Abstract
Mycotoxins are toxic secondary metabolites produced by certain filamentous fungi. The occurrence of mycotoxins in food and feed causes negative health impacts on both humans and animals. Clay binders, yeast cell walls, or antioxidant additives are the most widely used products for mycotoxin elimination to reduce their impact. Although conventional methods are constantly improving, current research trends are looking for innovative solutions. Nanotechnology approaches seem to be a promising, effective, and low-cost way to minimize the health effects of mycotoxins. This review aims to shed light on the critical knowledge gap in mycotoxin elimination by nanotechnology. There are three main strategies: mold inhibition, mycotoxin adsorption, and reducing the toxic effect via nanoparticles. One of the most promising methods is the use of carbon-based nanomaterials. Graphene has been shown to have a huge surface and high binding capacity for mycotoxins. Attention has also been drawn to polymeric nanoparticles; they could substitute adsorbents or enclose any substance, which would improve the health status of the organism. In light of these findings, this review gives new insights into possible future research that might overcome challenges associated with nanotechnology utilization for mycotoxin elimination from agricultural products.
Collapse
Affiliation(s)
- Pavel Horky
- Department of Animal Nutrition and Forage Production, Faculty of AgriSciences, Mendel University, 61300 Brno, Czech Republic.
| | - Sylvie Skalickova
- Department of Animal Nutrition and Forage Production, Faculty of AgriSciences, Mendel University, 61300 Brno, Czech Republic.
| | - Daria Baholet
- Department of Animal Nutrition and Forage Production, Faculty of AgriSciences, Mendel University, 61300 Brno, Czech Republic.
| | - Jiri Skladanka
- Department of Animal Nutrition and Forage Production, Faculty of AgriSciences, Mendel University, 61300 Brno, Czech Republic.
| |
Collapse
|
44
|
Tonigold M, Simon J, Estupiñán D, Kokkinopoulou M, Reinholz J, Kintzel U, Kaltbeitzel A, Renz P, Domogalla MP, Steinbrink K, Lieberwirth I, Crespy D, Landfester K, Mailänder V. Pre-adsorption of antibodies enables targeting of nanocarriers despite a biomolecular corona. NATURE NANOTECHNOLOGY 2018; 13:862-869. [PMID: 29915272 DOI: 10.1038/s41565-018-0171-6] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/21/2018] [Indexed: 05/17/2023]
Abstract
To promote drug delivery to exact sites and cell types, the surface of nanocarriers is functionalized with targeting antibodies or ligands, typically coupled by covalent chemistry. Once the nanocarrier is exposed to biological fluid such as plasma, however, its surface is inevitably covered with various biomolecules forming the protein corona, which masks the targeting ability of the nanoparticle. Here, we show that we can use a pre-adsorption process to attach targeting antibodies to the surface of the nanocarrier. Pre-adsorbed antibodies remain functional and are not completely exchanged or covered by the biomolecular corona, whereas coupled antibodies are more affected by this shielding. We conclude that pre-adsorption is potentially a versatile, efficient and rapid method of attaching targeting moieties to the surface of nanocarriers.
Collapse
Affiliation(s)
- Manuel Tonigold
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Johanna Simon
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | | | - Jonas Reinholz
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Ulrike Kintzel
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Patricia Renz
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Matthias P Domogalla
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kerstin Steinbrink
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | | | - Daniel Crespy
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Volker Mailänder
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
- Max Planck Institute for Polymer Research, Mainz, Germany.
| |
Collapse
|
45
|
Jamali Z, Khoobi M, Hejazi SM, Eivazi N, Abdolahpour S, Imanparast F, Moradi-Sardareh H, Paknejad M. Evaluation of targeted curcumin (CUR) loaded PLGA nanoparticles for in vitro photodynamic therapy on human glioblastoma cell line. Photodiagnosis Photodyn Ther 2018; 23:190-201. [PMID: 29969678 DOI: 10.1016/j.pdpdt.2018.06.026] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 05/16/2018] [Accepted: 06/29/2018] [Indexed: 02/08/2023]
Abstract
In this study, antibody-conjugated biodegradable polymeric nanoparticles were developed to enhance the photodaynamic efficiency of curcumin (CUR) on glioblastoma tumor cells. Poly (D, l-lactic-co-glycolic acid) nanoparticles (PLGA NPs) were synthesized and stabilized by polyvinyl alcohol (PVA). Poly(ethylene-alt-maleic anhydride) (PEMA) was used to provide carboxyl groups on the surface of NPs. The CUR or FITC (fluorescein isothiocyanate) was encapsulated in PLGA NPs using the nanoprecipitation method. The carboxylic groups on the surface of the PLGA NPs were covalently conjugated to the amino groups of a monoclonal antibody against EGFRvIII (A-EGFRvIII-f). The prepared NPs were fully characterized by Zetasizer, scanning electron microscope (SEM), differential scanning calorimetry (DSC), and Fourier transform infrared (FTIR), and then entrapment efficiency (EE), drug loading efficiency (DLE), CUR release, cell internalization, intrinsic cytotoxicity, and phototoxicity were evaluated. Furthermore, the effect of monoclonal antibody (MAb) on the tyrosine phosphorylation of EGFRvIII after photodynamic therapy (PDT) was assessed. The immunoreactivity of the antibody in MAb-PLGA NPs was preserved during the process of conjugation. The selective cellular internalization of MAb-PLGA NPs (FITC or CUR loaded) into the DKMG/EGFRvIII cells (EGFRvIII overexpressed human glioblastoma cell line) in comparison with DK-MGlow (human glioblastoma cell line with low level of EGFRvIII) was also confirmed. MAb-CUR-PLGA NPs were able to show more effective photodynamic toxicity (56% vs. 24%) on the DKMG/EGFRvIII cells compared to CUR-PLGA NPs. These results suggest that the anti-EGFRvIII MAb-CUR-PLGA NPs have potential of targeted drug delivery system for PDT in the overexpressed EGFRvIII tumor cells.
Collapse
Affiliation(s)
- Zahra Jamali
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Khoobi
- Nanobiomaterials Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Marjaneh Hejazi
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Bio Optical Imaging Group, Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Eivazi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeideh Abdolahpour
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Imanparast
- Department of Medical Biochemistry, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Hemen Moradi-Sardareh
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Paknejad
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Alibakhshi A, Abarghooi Kahaki F, Ahangarzadeh S, Yaghoobi H, Yarian F, Arezumand R, Ranjbari J, Mokhtarzadeh A, de la Guardia M. Targeted cancer therapy through antibody fragments-decorated nanomedicines. J Control Release 2017; 268:323-334. [DOI: 10.1016/j.jconrel.2017.10.036] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/22/2017] [Accepted: 10/24/2017] [Indexed: 01/10/2023]
|
47
|
Fogli S, Montis C, Paccosi S, Silvano A, Michelucci E, Berti D, Bosi A, Parenti A, Romagnoli P. Inorganic nanoparticles as potential regulators of immune response in dendritic cells. Nanomedicine (Lond) 2017. [PMID: 28635380 DOI: 10.2217/nnm-2017-0061] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
AIM The spontaneous adsorption of proteins on nanoparticles (NPs) in biological media is exploited to prepare complexes of NPs and proteins from cancer cells' lysates for application in cancer immunotherapy. MATERIALS & METHODS Gold (Au) and silica NPs were synthesized, incubated with cancer cells' lysates and characterized. Dendritic cells (DCs) were challenged with protein-coated NPs, their maturation, viability and morphology were evaluated and lymphocytes T proliferation was determined. RESULTS Silica and Au NPs bound different pools of biomolecules from lysates, and are therefore promising selective carriers for antigens. When incubated with immature DCs, NPs were efficiently endocytosed without cytotoxicity. Finally, protein-coated AuNPs promoted DC maturation and DC-mediated lymphocyte proliferation, at variance with lysate alone and protein-coated silica NPs, that did not promote DCs maturation. CONCLUSION These results demonstrate that the spontaneous formation of protein coronas on NPs represents a possible approach to fast, easy, cost-effective DCs stimulation.
Collapse
Affiliation(s)
- Silvia Fogli
- Department of Chemistry 'Ugo Schiff,' University of Florence, and CSGI, via della Lastruccia 3, 50019 Sesto Fiorentino, FI, Italy
| | - Costanza Montis
- Department of Chemistry 'Ugo Schiff,' University of Florence, and CSGI, via della Lastruccia 3, 50019 Sesto Fiorentino, FI, Italy
| | - Sara Paccosi
- Department of Health Sciences, Clinical Pharmacology & Oncology Section, University of Florence, Viale Pieraccini 6, 50139, FI, Italy
| | - Angela Silvano
- Department of Experimental & Clinical Medicine, University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, FI, Italy
| | - Elena Michelucci
- Mass Spectrometry Center (CISM), University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, FI, Italy
| | - Debora Berti
- Department of Chemistry 'Ugo Schiff,' University of Florence, and CSGI, via della Lastruccia 3, 50019 Sesto Fiorentino, FI, Italy
| | - Alberto Bosi
- Department of Experimental & Clinical Medicine, University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, FI, Italy
| | - Astrid Parenti
- Department of Health Sciences, Clinical Pharmacology & Oncology Section, University of Florence, Viale Pieraccini 6, 50139, FI, Italy
| | - Paolo Romagnoli
- Department of Experimental & Clinical Medicine, University of Florence, via Ugo Schiff 6, 50019 Sesto Fiorentino, FI, Italy
| |
Collapse
|
48
|
Karacivi M, Sumer Bolu B, Sanyal R. Targeting to the Bone: Alendronate-Directed Combretastatin A-4 Bearing Antiangiogenic Polymer-Drug Conjugates. Mol Pharm 2017; 14:1373-1383. [PMID: 28358515 DOI: 10.1021/acs.molpharmaceut.6b01173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Selective targeting of tumor site with chemotherapeutic agents appears to be one of the most effective methods to address many of the problems encountered with conventional chemotherapy. In this work, poly(oligoethylene glycol)methacrylate (POEGMA) based bone-targeting polymers bearing an antiangiogenic drug combretastatin A4 (CA4) were synthesized using free radical polymerization. Targeted and nontargeting copolymers were evaluated for their bone targeting efficiency, cytotoxicities against endothelial cells, namely, HUVECs and U2-OS and Saos-2 cancerous cell lines, as well as their antiangiogenic activity against endothelial cell tube formation by HUVECs. It is observed that the drug conjugated polymers conjugated with the bisphosphonate groups containing drug alendronate (ALN) have remarkably high affinity for bone mineral when compared to the polymer-drug conjugates devoid of the bisphosphonate groups. Both targeted and nontargeted polymer-drug conjugates show a sustained drug release in rat plasma with an overall release of 80-93% over 5 days. In vitro studies revealed high levels of cytotoxicity of the polymer-drug conjugates against HUVECs and U2-OS, and moderate cytotoxicity toward Saos-2. Importantly, the CA4 conjugated copolymers displayed excellent level of antiangiogenic activity as deduced from in vitro endothelial cell tube formation assay using HUVECs. Overall, a novel bone-targeting antiangiogenic polymer-drug conjugate that can be further elaborated to carry additional anticancer drugs is disclosed.
Collapse
Affiliation(s)
- Merve Karacivi
- Department of Chemistry and ‡Center for Life Science and Technologies, Bogazici University , Istanbul 34342, Turkey
| | - Burcu Sumer Bolu
- Department of Chemistry and ‡Center for Life Science and Technologies, Bogazici University , Istanbul 34342, Turkey
| | - Rana Sanyal
- Department of Chemistry and ‡Center for Life Science and Technologies, Bogazici University , Istanbul 34342, Turkey
| |
Collapse
|
49
|
Velpurisiva P, Gad A, Piel B, Jadia R, Rai P. Nanoparticle Design Strategies for Effective Cancer Immunotherapy. JOURNAL OF BIOMEDICINE (SYDNEY, NSW) 2017; 2:64-77. [PMID: 28503405 PMCID: PMC5426812 DOI: 10.7150/jbm.18877] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cancer immunotherapy is a rapidly evolving and paradigm shifting treatment modality that adds a strong tool to the collective cancer treatment arsenal. It can be effective even for late stage diagnoses and has already received clinical approval. Tumors are known to not only avoid immune surveillance but also exploit the immune system to continue local tumor growth and metastasis. Because of this, most immunotherapies, particularly those directed against solid cancers, have thus far only benefited a small minority of patients. Early clinical substantiation lends weight to the claim that cancer immunotherapies, which are adaptive and enduring treatment methods, generate much more sustained and robust anticancer effects when they are effectively formulated in nanoparticles or scaffolds than when they are administered as free drugs. Engineering cancer immunotherapies using nanomaterials is, therefore, a very promising area worthy of further consideration and investigation. This review focuses on the recent advances in cancer immunoengineering using nanoparticles for enhancing the therapeutic efficacy of a diverse range of immunotherapies. The delivery of immunostimulatory agents to antitumor immune cells, such as dendritic or antigen presenting cells, may be a far more efficient tactic to eradicate tumors than delivery of conventional chemotherapeutic and cytotoxic drugs to cancer cells. In addition to its immense therapeutic potential, immunoengineering using nanoparticles also provides a valuable tool for unearthing and understanding the basics of tumor biology. Recent research using nanoparticles for cancer immunotherapy has demonstrated the advantage of physicochemical manipulation in improving the delivery of immunostimulatory agents. In vivo studies have tested a range of particle sizes, mostly less than 300 nm, and particles with both positive and negative zeta potentials for various applications. Material composition and surface modifications have been shown to contribute significantly in selective targeting, efficient delivery and active stimulation of immune system targets. Thus, these investigations, including a wide array of nanoparticles for cancer immunotherapy, substantiate the employment of nanocarriers for efficacious cancer immunotherapies.
Collapse
Affiliation(s)
- Praveena Velpurisiva
- Department of Biomedical Engineering and Biotechnology, University of Massachusetts Lowell, USA
| | - Aniket Gad
- Department of Biomedical Engineering and Biotechnology, University of Massachusetts Lowell, USA
| | - Brandon Piel
- Department of Chemical Engineering, University of Massachusetts Lowell, USA
| | - Rahul Jadia
- Department of Biomedical Engineering and Biotechnology, University of Massachusetts Lowell, USA
| | - Prakash Rai
- Department of Chemical Engineering, University of Massachusetts Lowell, USA
| |
Collapse
|
50
|
Centelles MN, Wright M, Gedroyc W, Thanou M. Focused ultrasound induced hyperthermia accelerates and increases the uptake of anti-HER-2 antibodies in a xenograft model. Pharmacol Res 2016; 114:144-151. [PMID: 27771465 DOI: 10.1016/j.phrs.2016.10.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 10/05/2016] [Accepted: 10/18/2016] [Indexed: 01/07/2023]
Abstract
Image guided drug delivery has gained significant attention during the last few years. Labelling nanoparticles or macromolecules and monitoring their fate in the body provides information that can be used to modulate their biodistribution and improve their pharmacokinetics. In this study we label antibodies and monitor their distribution in the tumours post intravenous injection. Using Focused Ultrasound (FUS, a non-invasive method of hyperthermia) we increase the tumour temperature to 42°C for a short period of time (3-5min) and we observe an increased accumulation of labelled antibody. Repetition of focused ultrasound induced hyperthermic treatment increased still further the accumulation of the antibodies in the tumour. This treatment also augmented the accumulation of other macromolecules non-specific to the tumour, such as IgG and albumin. These effects may be used to enhance the therapeutic efficiency of antibodies and/or targeted nanoparticles.
Collapse
Affiliation(s)
| | - Michael Wright
- Institute of Pharmaceutical Science, King's College London, UK
| | | | - Maya Thanou
- Institute of Pharmaceutical Science, King's College London, UK.
| |
Collapse
|