1
|
Izadiyan Z, Misran M, Kalantari K, Webster TJ, Kia P, Basrowi NA, Rasouli E, Shameli K. Advancements in Liposomal Nanomedicines: Innovative Formulations, Therapeutic Applications, and Future Directions in Precision Medicine. Int J Nanomedicine 2025; 20:1213-1262. [PMID: 39911259 PMCID: PMC11794392 DOI: 10.2147/ijn.s488961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 01/01/2025] [Indexed: 02/07/2025] Open
Abstract
Liposomal nanomedicines have emerged as a pivotal approach for the treatment of various diseases, notably cancer and infectious diseases. This manuscript provides an in-depth review of recent advancements in liposomal formulations, highlighting their composition, targeted delivery strategies, and mechanisms of action. We explore the evolution of liposomal products currently in clinical trials, emphasizing their potential in addressing diverse medical challenges. The integration of immunotherapeutic agents within liposomes marks a paradigm shift, enabling the design of 'immuno-modulatory hubs' capable of orchestrating precise immune responses while facilitating theranostic applications. The recent COVID-19 pandemic has accelerated research in liposomal-based vaccines and antiviral therapies, underscoring the need for improved delivery mechanisms to overcome challenges like rapid clearance and organ toxicity. Furthermore, we discuss the potential of "smart" liposomes, which can respond to specific disease microenvironments, enhancing treatment efficacy and precision. The integration of artificial intelligence and machine learning in optimizing liposomal designs promises to revolutionize personalized medicine, paving the way for innovative strategies in disease detection and therapeutic interventions. This comprehensive review underscores the significance of ongoing research in liposomal technologies, with implications for future clinical applications and enhanced patient outcomes.
Collapse
Affiliation(s)
- Zahra Izadiyan
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Misni Misran
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Katayoon Kalantari
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Thomas J Webster
- Biomedical Engineering, Hebei University of Technology, Tianjin, People’s Republic of China
- School of Engineering, Saveetha University, Chennai, India
| | - Pooneh Kia
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | | - Elisa Rasouli
- Department of Electrical and Electronics Engineering, Nanyang Technological University, Nanyang, Singapore
| | - Kamyar Shameli
- School of Medicine, Institute of Virology, Technical University of Munich, Munich, Germany
| |
Collapse
|
2
|
Topçu BT, Bozdağ Pehlivan S, Akdağ Y, Mut M, Öner L. Antibody Conjugated Nano-Enabled Drug Delivery Systems Against Brain Tumors. J Pharm Sci 2024; 113:1455-1469. [PMID: 38555997 DOI: 10.1016/j.xphs.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024]
Abstract
The use of antibody-conjugated nanoparticles for brain tumor treatment has gained significant attention in recent years. Nanoparticles functionalized with anti-transferrin receptor antibodies have shown promising results in facilitating nanoparticle uptake by endothelial cells of brain capillaries and post-capillary venules. This approach offers a potential alternative to the direct conjugation of biologics to antibodies. Furthermore, studies have demonstrated the potential of antibody-conjugated nanoparticles in targeting brain tumors, as evidenced by the specific binding of these nanoparticles to brain cancer cells. Additionally, the development of targeted nanoparticles designed to transcytoses the blood-brain barrier (BBB) to deliver small molecule drugs and therapeutic antibodies to brain metastases holds promise for brain tumor treatment. While the use of nanoparticles as a delivery method for brain cancer treatment has faced challenges, including the successful delivery of nanoparticles to malignant brain tumors due to the presence of the BBB and infiltrating cancer cells in the normal brain, recent advancements in nanoparticle-mediated drug delivery systems have shown potential for enhancing the efficacy of brain cancer therapy. Moreover, the development of brain-penetrating nanoparticles capable of distributing over clinically relevant volumes when administered via convection-enhanced delivery presents a promising strategy for improving drug delivery to brain tumors. In conclusion, the use of antibody-conjugated nanoparticles for brain tumor treatment shows great promise in overcoming the challenges associated with drug delivery to the brain. By leveraging the specific targeting capabilities of these nanoparticles, researchers are making significant strides in developing effective and targeted therapies for brain tumors.
Collapse
Affiliation(s)
- Beril Taş Topçu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University 06100, Ankara, Turkey
| | - Sibel Bozdağ Pehlivan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University 06100, Ankara, Turkey.
| | - Yagmur Akdağ
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University 06100, Ankara, Turkey
| | - Melike Mut
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22903, USA
| | - Levent Öner
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University 06100, Ankara, Turkey
| |
Collapse
|
3
|
Fidan Y, Muçaj S, Timur SS, Gürsoy RN. Recent advances in liposome-based targeted cancer therapy. J Liposome Res 2024; 34:316-334. [PMID: 37814217 DOI: 10.1080/08982104.2023.2268710] [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: 05/13/2023] [Accepted: 09/29/2023] [Indexed: 10/11/2023]
Abstract
Nano-drug delivery systems have opened new pathways for tumor treatment by overcoming some of the limitations of conventional drugs, such as physiological degradation, short half-life, and rapid release. Liposomes are promising nanocarrier systems due to their biocompatibility, low toxicity, and high inclusivity, as well as their enhanced drug bioavailability. Various strategies for active targeting of liposomal formulations have been investigated to achieve the highest drug efficacy. This review aims to summarize current developments in novel liposomal formulations, particularly ligand-targeted liposomes (such as folate, transferrin, hyaluronic acid, antibodies, aptamer, and peptide, etc.) used for the therapy of various cancers and provide an insight on the challenges and future of liposomes for scientists and pharmaceutical companies.
Collapse
Affiliation(s)
- Yeliz Fidan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Stela Muçaj
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Selin Seda Timur
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - R Neslihan Gürsoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
4
|
Nair M, Chandra A, Krishnan A, Chandra A, Basha R, Orimoloye H, Raut S, Gayathri V, Mudgapalli VV, Vishwanatha JK. Protein and peptide nanoparticles for drug delivery applications. NANOSTRUCTURED MATERIALS FOR BIOMEDICAL APPLICATIONS 2024:339-404. [DOI: 10.1016/b978-0-323-90838-2.00011-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Wang T, Wu M, Cao L, Liu B. Organic functional substance engineered living materials for biomedical applications. Biomaterials 2023; 301:122248. [PMID: 37487360 DOI: 10.1016/j.biomaterials.2023.122248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/09/2023] [Accepted: 07/16/2023] [Indexed: 07/26/2023]
Abstract
Modifying living materials with organic functional substances (OFS) is a convenient and effective strategy to control and monitor the transport, engraftment, and secretion processes in living organisms. OFSs, including small organic molecules and organic polymers, own the merit of design flexibility, satisfying performance, and excellent biocompatibility, which allow for living materials functionalization to realize real-time sensing, controlled drug release, enhanced biocompatibility, accurate diagnosis, and precise treatment. In this review, we discuss the different principles of OFS modification on living materials and demonstrate the applications of engineered living materials in health monitoring, drug delivery, wound healing, and tissue regeneration.
Collapse
Affiliation(s)
- Tongtong Wang
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China; Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585, Singapore
| | - Min Wu
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China.
| | - Lei Cao
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China; Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585, Singapore
| | - Bin Liu
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China; Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585, Singapore.
| |
Collapse
|
6
|
Functionalization of Nanosystems in Cancer Treatment. Cancer Nanotechnol 2023. [DOI: 10.1007/978-3-031-17831-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
7
|
Kamyab H, Chelliapan S, Tavakkoli O, Mesbah M, Bhutto JK, Khademi T, Kirpichnikova I, Ahmad A, ALJohani AA. A review on carbon-based molecularly-imprinted polymers (CBMIP) for detection of hazardous pollutants in aqueous solutions. CHEMOSPHERE 2022; 308:136471. [PMID: 36126738 DOI: 10.1016/j.chemosphere.2022.136471] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/30/2022] [Accepted: 09/12/2022] [Indexed: 06/15/2023]
Abstract
This article discusses the unique properties and performance of carbon-based molecularly-imprinted polymers (MIPs) for detecting hazardous pollutants in aqueous solutions. Although MIPs have several advantages such as specific recognition sites, selectivity, and stability, they suffer from a series of drawbacks, including loss of conductivity, electrocatalytic activity, and cost, which limit their use in various fields. Carbon-based MIPs, which utilize carbon electrodes, carbon nanoparticles, carbon dots, carbon nanotubes, and graphene substrates, have been the focus of research in recent years to enhance their properties and remove their weaknesses as much as possible. These carbon-based nanomaterials have excellent sensitivity and specificity for molecular identification. As a result, they have been widely used in various applications, such as assessing the environmental, biological, and food samples. This article examines the growth of carbon-based MIPs and their environmental applications.
Collapse
Affiliation(s)
- Hesam Kamyab
- Malaysia-Japan International Institute of Technology, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia; Department of Biomaterials, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 600 077, India; Department of Electric Power Stations, Network and Supply Systems, South Ural State University (National Research University), 76 Prospekt Lenina, 454080, Chelyabinsk, Russian Federation.
| | - Shreeshivadasan Chelliapan
- Engineering Department, Razak Faculty of Technology & Informatics, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia
| | - Omid Tavakkoli
- Department of Petroleum Engineering, Faculty of Chemical and Energy Engineering, Universiti Teknologi Malaysia, 81310 Skudai, Johor Bahru, Malaysia
| | - Mohsen Mesbah
- Engineering Department, Razak Faculty of Technology & Informatics, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia.
| | - Javed Khan Bhutto
- Department of Electrical Engineering, College of Engineering, King Khalid University, Abha, Saudi Arabia
| | - Tayebeh Khademi
- Azman Hashim International Business School (AHIBS), Universiti Teknologi Malaysia, 81310, Skudai, Johor, Malaysia
| | - Irina Kirpichnikova
- Department of Electric Power Stations, Network and Supply Systems, South Ural State University (National Research University), 76 Prospekt Lenina, 454080, Chelyabinsk, Russian Federation
| | - Akil Ahmad
- Chemistry Department, College of Science and Humanities, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Anas Ayesh ALJohani
- Department of Electrical Engineering, College of Engineering, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
8
|
Hsieh HH, Kuo WY, Lin JJ, Chen HS, Hsu HJ, Wu CY. Tumor-Targeting Ability of Novel Anti-Prostate-Specific Membrane Antigen Antibodies. ACS OMEGA 2022; 7:31529-31537. [PMID: 36092556 PMCID: PMC9454275 DOI: 10.1021/acsomega.2c04230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/12/2022] [Indexed: 06/15/2023]
Abstract
Patients with prostate-specific membrane antigen (PSMA)-positive tumors can benefit from PSMA-targeted therapy; thus, we have constructed a phage-displayed synthetic antibody library for the production of novel PSMA antibodies with superior PSMA-targeting ability, favoring clinical management. The binding affinities of anti-PSMA antibodies were verified by an enzyme-linked immunosorbent assay (ELISA). Several in vitro and in vivo experiments, including cellular uptake, internalization, and cytotoxicity studies, micro single photon emission computed tomography (microSPECT)/CT, and biodistribution studies, were performed to select the most promising antibody among six different antibodies. The results showed the target affinities of our antibodies in the ELISA assays (7A, 8C, 8E, and 11A) were comparable to the existing antibodies (J591). The half-maximal effective concentrations of 7A, 8C, 8E, 11A, and J591 were 2.95, 6.64, 5.50, 2.08, and 4.79, respectively. The radiochemical yield of 111In-labeled antibodies ranged from 30% to 50% with high radiochemical purity (>90%). In the cellular uptake studies, the accumulated radioactivity of 111In-J591, 111In-7A, and 111In-11A increased over time. The internalized percentage of 111In-11A was the highest (32.14% ± 2.06%) at 48 h after incubation, whereas that of 111In-J591 peaked at 22.43% ± 4.38% at 24 h and dropped to 13.52% ± 3.03% at 48 h postincubation. Twenty-four hours after injection, radioactivity accumulation appeared in the LNCaP xenografts of the mice injected with 111In-11A, 111In-8E, 111In-7A, and 111In-J591 but not in the xenografts of the 111In-8C-injected group. Marked liver uptake was noticed in all groups except the 111In-11A-injected group. Moreover, the killing effect of 177Lu-11A was superior to that of 177Lu-J591 at low concentrations. In conclusion, we successfully demonstrated that 11A IgG owned the most optimal biological characteristics among several new anti-PSMA antibodies and it can be an excellent PSMA-targeting component for the clinical use.
Collapse
Affiliation(s)
- Hsin-Hua Hsieh
- Department
of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei 112, Taiwan
| | - Wei-Ying Kuo
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Jia-Jia Lin
- Department
of Nuclear Medicine, New Taipei Municipal
TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation), New Taipei City 236, Taiwan
| | - Hong-Sen Chen
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Hung-Ju Hsu
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chun-Yi Wu
- Department
of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei 112, Taiwan
| |
Collapse
|
9
|
Jin Y, Edalatian Zakeri S, Bahal R, Wiemer AJ. New Technologies Bloom Together for Bettering Cancer Drug Conjugates. Pharmacol Rev 2022; 74:680-711. [PMID: 35710136 PMCID: PMC9553120 DOI: 10.1124/pharmrev.121.000499] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Drug conjugates, including antibody-drug conjugates, are a step toward realizing Paul Ehrlich's idea from over 100 years ago of a "magic bullet" for cancer treatment. Through balancing selective targeting molecules with highly potent payloads, drug conjugates can target specific tumor microenvironments and kill tumor cells. A drug conjugate consists of three parts: a targeting agent, a linker, and a payload. In some conjugates, monoclonal antibodies act as the targeting agent, but new strategies for targeting include antibody derivatives, peptides, and even small molecules. Linkers are responsible for connecting the payload to the targeting agent. Payloads impact vital cellular processes to kill tumor cells. At present, there are 12 antibody-drug conjugates on the market for different types of cancers. Research on drug conjugates is increasing year by year to solve problems encountered in conjugate design, such as tumor heterogeneity, poor circulation, low drug loading, low tumor uptake, and heterogenous expression of target antigens. This review highlights some important preclinical research on drug conjugates in recent years. We focus on three significant areas: improvement of antibody-drug conjugates, identification of new conjugate targets, and development of new types of drug conjugates, including nanotechnology. We close by highlighting the critical barriers to clinical translation and the open questions going forward. SIGNIFICANCE STATEMENT: The development of anticancer drug conjugates is now focused in three broad areas: improvements to existing antibody drug conjugates, identification of new targets, and development of new conjugate forms. This article focuses on the exciting preclinical studies in these three areas and advances in the technology that improves preclinical development.
Collapse
Affiliation(s)
- Yiming Jin
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut
| | | | - Raman Bahal
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut
| | - Andrew J Wiemer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
10
|
Nadukkandy AS, Ganjoo E, Singh A, Dinesh Kumar L. Tracing New Landscapes in the Arena of Nanoparticle-Based Cancer Immunotherapy. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.911063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Over the past two decades, unique and comprehensive cancer treatment has ushered new hope in the holistic management of the disease. Cancer immunotherapy, which harnesses the immune system of the patient to attack the cancer cells in a targeted manner, scores over others by being less debilitating compared to the existing treatment strategies. Significant advancements in the knowledge of immune surveillance in the last few decades have led to the development of several types of immune therapy like monoclonal antibodies, cancer vaccines, immune checkpoint inhibitors, T-cell transfer therapy or adoptive cell therapy (ACT) and immune system modulators. Intensive research has established cancer immunotherapy to be a safe and effective method for improving survival and the quality of a patient’s life. However, numerous issues with respect to site-specific delivery, resistance to immunotherapy, and escape of cancer cells from immune responses, need to be addressed for expanding and utilizing this therapy as a regular mode in the clinical treatment. Development in the field of nanotechnology has augmented the therapeutic efficiency of treatment modalities of immunotherapy. Nanocarriers could be used as vehicles because of their advantages such as increased surface areas, targeted delivery, controlled surface and release chemistry, enhanced permeation and retention effect, etc. They could enhance the function of immune cells by incorporating immunomodulatory agents that influence the tumor microenvironment, thus enabling antitumor immunity. Robust validation of the combined effect of nanotechnology and immunotherapy techniques in the clinics has paved the way for a better treatment option for cancer than the already existing procedures such as chemotherapy and radiotherapy. In this review, we discuss the current applications of nanoparticles in the development of ‘smart’ cancer immunotherapeutic agents like ACT, cancer vaccines, monoclonal antibodies, their site-specific delivery, and modulation of other endogenous immune cells. We also highlight the immense possibilities of using nanotechnology to accomplish leveraging the coordinated and adaptive immune system of a patient to tackle the complexity of treating unique disease conditions and provide future prospects in the field of cancer immunotherapy.
Collapse
|
11
|
Janani B, Vijayakumar M, Priya K, Kim JH, Prabakaran DS, Shahid M, Al-Ghamdi S, Alsaidan M, Othman Bahakim N, Hassan Abdelzaher M, Ramesh T. EGFR-Based Targeted Therapy for Colorectal Cancer—Promises and Challenges. Vaccines (Basel) 2022; 10:vaccines10040499. [PMID: 35455247 PMCID: PMC9030067 DOI: 10.3390/vaccines10040499] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/05/2023] Open
Abstract
Colorectal carcinoma (CRC) is the most lethal and common form of cancer in the world. It was responsible for almost 881,000 cancer deaths in 2018. Approximately 25% of cases are diagnosed at advanced stages with metastasis—this poses challenges for effective surgical control and future tumor-related mortality. There are numerous diagnostic methods that can be used to reduce the risk of colorectal carcinoma. Among these, targeted nanotherapy aims to eliminate the tumor and any metastasis. Active targeting can increase the effectiveness and quantity of drugs delivered to the target site. Antibodies that target overexpressed receptors on cell surfaces and indicators are coupled with drug-loaded carriers. The major target receptors of chemotherapeutic drugs delivery include VEGFR, EGFR, FGFR, HER2, and TGF. On account of its major and diverse roles in cancer, it is important to target EGFR in particular for better tumor selection, as EGFR is overexpressed in 25 to 82% of colorectal carcinoma cases. The EGFR monoclonal immunoglobulins cetuximab/panitumumab can thus be used to treat colorectal cancer. This review examines carriers that contain cetuximab-conjugated therapeutic drugs as well as their efficacy in anticancer activities.
Collapse
Affiliation(s)
- Balakarthikeyan Janani
- Department of Biochemistry, PSG College of Arts and Science (Autonomous), Bharathiar University, Coimbatore 641014, Tamil Nadu, India;
| | - Mayakrishnan Vijayakumar
- Department of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, 209 Neugdong-ro, Gwangjin-gu, Seoul 05006, Korea; (M.V.); (J.H.K.)
| | - Kannappan Priya
- Department of Biochemistry, PSG College of Arts and Science (Autonomous), Bharathiar University, Coimbatore 641014, Tamil Nadu, India;
- Correspondence: (K.P.); (T.R.)
| | - Jin Hee Kim
- Department of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, 209 Neugdong-ro, Gwangjin-gu, Seoul 05006, Korea; (M.V.); (J.H.K.)
| | - D. S. Prabakaran
- Department of Radiation Oncology, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju 28644, Korea;
- Department of Biotechnology, Ayya Nadar Janaki Ammal College (Autonomous), Srivilliputhur Main Road, Sivakasi 626124, Tamil Nadu, India
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (M.S.); (N.O.B.); (M.H.A.)
| | - Sameer Al-Ghamdi
- Family and Community Medicine Department, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Mohammed Alsaidan
- Internal Medicine Department, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Nasraddin Othman Bahakim
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (M.S.); (N.O.B.); (M.H.A.)
| | - Mohammad Hassan Abdelzaher
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (M.S.); (N.O.B.); (M.H.A.)
- Department of Medical Biochemistry, Faculty of Medicine, Al-Azhar University (Assiut Branch), Assiut 71515, Egypt
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (M.S.); (N.O.B.); (M.H.A.)
- Correspondence: (K.P.); (T.R.)
| |
Collapse
|
12
|
Wang Z, Wang P, Song H, Chen Z. Dynamic wetting behavior of nanofluid droplet on a vertically vibrating surface: A molecular dynamics study. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2021.118360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
13
|
Fatima SW, Khare SK. Benefits and challenges of antibody drug conjugates as novel form of chemotherapy. J Control Release 2021; 341:555-565. [PMID: 34906604 DOI: 10.1016/j.jconrel.2021.12.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022]
Abstract
Antibody drug conjugates (ADCs) are an emerging therapeutic modality for targeted cancer treatment. They represent the unique amalgamation of chemotherapy and immunotherapy. ADCs comprise of monoclonal antibodies linked with drugs (payloads) through a chemical linker designed to deliver the cytotoxic moiety to the cancer cells. The present paper is a review of recent clinical advances of each component of ADCs (antibody/linker/payload) and how the individual component influences the activity of ADCs. The review discusses opportunities for improving ADCs efficiency and ways to have a better antibody-based molecular platform, which could substantially increase chemotherapy outcomes. This review casts an outlook on how ADCs enhancement in terms of their pharmacokinetics, therapeutic indexes and safety profiles can overcome the prevailing challenges like drug resistance in cancer treatment. A novel strategy of augmenting antibodies with nanoparticles anticipates a huge success in terms of targeted delivery of drugs in several diseases. Antibody conjugated nanoparticles (ACNPs) are a very promising strategy for the cutting-edge development of chemo/immunotherapies for efficient delivery of payloads at the targeted cancer cells. The avenues of a high drug to antibody ratio (DAR) owing to the selection of broad chemotherapy payloads, regulating drug release eliciting higher avidity of ACNPs over ADCs will be the modern immunotherapeutics. ACNPs carry immense potential to mark a paradigm shift in cancer chemotherapy that may be a substitute for ADCs.
Collapse
Affiliation(s)
- Syeda Warisul Fatima
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Sunil K Khare
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, New Delhi 110016, India.
| |
Collapse
|
14
|
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
|
15
|
Tian H, Huang Y, He J, Zhang M, Ni P. CD147 Monoclonal Antibody Targeted Reduction-Responsive Camptothecin Polyphosphoester Nanomedicine for Drug Delivery in Hepatocellular Carcinoma Cells. ACS APPLIED BIO MATERIALS 2021; 4:4422-4431. [PMID: 35006854 DOI: 10.1021/acsabm.1c00177] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the treatment of tumor-targeted small-molecule anti-cancer drugs, antibody-mediated therapies, especially for antibody-drug conjugates (ADCs), have revealed great latent force. However, the therapeutic drugs provided by ADCs possess limitation. Considering that the combination of antibodies and nano-drugs can broaden their applicability in the field of tumor treatment, herein, we developed an antibody conjugated polymeric prodrug nanoparticles SAE-PEG-b-PBYP-ss-CPT for targeted camptothecin (CPT) delivery to liver tumor cells. The diblock copolymer was composed of PEG and biodegradable polyphosphoester (PBYP) containing alkynyl groups in the side chain. A derivative of CPT (CPT-ss-N3) was bonded to the PBYP via "click" reaction. The diethyl squarate (SAE) in the terminal of PEG chain was used as a functional group to bond with CD147 monoclonal antibody (CD147 mAb). The particle size and size distribution of the both nanoparticles, with antibody binding (namely CD147-CPT NPs) and without antibody (abbreviated as CPT-loaded NPs), were measured by dynamic light scattering (DLS). The morphologies of both two kinds of nanoparticles were observed by transmission electron microscope (TEM). The results of X-ray photoelectron spectroscopy (XPS) showed that CD147 mAb had been coupled to the surface of CPT-loaded NPs. Endocytosis test indicated that CD147-CPT NPs had higher uptake rate and accumulation in HepG2 cells than those of CPT-loaded NPs without antibodies, due to CD147 mAb can specifically bind to CD147 protein overexpressed in HepG2 cells. We establish a method to bond monoclonal antibodies to anti-cancer polymeric prodrugs, and endow biodegradable polymeric prodrugs with precise targeting functions to liver cancer cells.
Collapse
Affiliation(s)
- Hongrui Tian
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou 215123, China
| | - Yakui Huang
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou 215123, China
| | - Jinlin He
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou 215123, China
| | - Mingzu Zhang
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou 215123, China
| | - Peihong Ni
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou 215123, China
| |
Collapse
|
16
|
Design of liposomes as drug delivery system for therapeutic applications. Int J Pharm 2021; 601:120571. [PMID: 33812967 DOI: 10.1016/j.ijpharm.2021.120571] [Citation(s) in RCA: 487] [Impact Index Per Article: 121.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/25/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022]
Abstract
Liposomes are spherical vesicles consisting of one or more concentric phospholipid bilayers enclosing an aqueous core. Being both nontoxic and biodegradable, liposomes represent a powerful delivery system for several drugs. They have improved the therapeutic efficacy of drugs through stabilizing compounds, overcoming obstacles to cellular and tissue uptake and increasing drug biodistribution to target sites in vivo, while minimizing systemic toxicity. This review offers an overview of liposomes, thought the exploration of their key fundamentals. Initially, the main design aspects to obtain a successful liposomal formulation were addressed, following the techniques for liposome production and drug loading. Before application, liposomes required an extensive characterization to assurance in vitro and in vivo performance. Thus, several properties to characterize liposomes were explored, such as size, polydispersity index, zeta potential, shape, lamellarity, phase behavior, encapsulation efficiency, and in vitro drug release. Topics related with liposomal functionalization and effective targeting strategies were also addressed, as well as stability and some limitations of liposomes. Finally, this review intends to explore the current market liposomes used as a drug delivery system in different therapeutic applications.
Collapse
|
17
|
Mirzavi F, Barati M, Soleimani A, Vakili-Ghartavol R, Jaafari MR, Soukhtanloo M. A review on liposome-based therapeutic approaches against malignant melanoma. Int J Pharm 2021; 599:120413. [PMID: 33667562 DOI: 10.1016/j.ijpharm.2021.120413] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 01/14/2023]
Abstract
Melanoma is a highly aggressive form of skin cancer with a very poor prognosis and excessive resistance to current conventional treatments. Recently, the application of the liposomal delivery system in the management of skin melanoma has been widely investigated. Liposomal nanocarriers are biocompatible and less toxic to host cells, enabling the efficient and safe delivery of different therapeutic agents into the tumor site and further promoting their antitumor activities. Therefore, the liposomal delivery system effectively increases the success of current melanoma therapies and overcomes resistance. In this review, we present an overview of liposome-based targeted drug delivery methods and highlight recent advances towards the development of liposome-based carriers for therapeutic genes. We also discuss the new insights regarding the efficacy and clinical significance of combinatorial treatment of liposomal formulations with immunotherapy and conventional therapies in melanoma patients for a better understanding and successfully managing cancer.
Collapse
Affiliation(s)
- Farshad Mirzavi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Barati
- Department of Medical Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Anvar Soleimani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Roghayyeh Vakili-Ghartavol
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
18
|
Taneja P, Sharma S, Sinha VB, Yadav AK. Advancement of nanoscience in development of conjugated drugs for enhanced disease prevention. Life Sci 2021; 268:118859. [PMID: 33358907 DOI: 10.1016/j.lfs.2020.118859] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/28/2020] [Accepted: 12/04/2020] [Indexed: 12/26/2022]
Abstract
Nanoscience and nanotechnology is a recently emerging and rapid developing field of science and has also been explored in the fields of Biotechnology and Medicine. Nanoparticles are being used as tools for diagnostic purposes and as a medium for the delivery of therapeutic agents to the specific targeted sites under controlled conditions. The physicochemical properties of these nanoparticles give them the ability to treat various chronic human diseases by site specific drug delivery and to use in diagnosis, biosensing and bioimaging devices, and implants. According to the type of materials used nanoparticles can be classified as organic (micelles, liposomes, nanogels and dendrimers) and inorganic (including gold nanoparticles (GNPs), super-paramagnetic iron oxide nanomaterials (SPIONs), quantum dots (QDs), and paramagnetic lanthanide ions). Different types of nanoparticle are being used in conjugation with various types of biomoities (such as peptide, lipids, antibodies, nucleotides, plasmids, ligands and polysaccharides) to form nanoparticle-drug conjugates which has enhanced capacity of drug delivery at targeted sites and hence improved disease treatment and diagnosis. In this study, the summary of various types of nanoparticle-drug conjugates that are being used along with their mechanism and applications are included. In addition, the various nanoparticle-drug conjugates which are being used and which are under clinical studies along with their future opportunities and challenges are also discussed in this review.
Collapse
Affiliation(s)
- Pankaj Taneja
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India.
| | - Sonali Sharma
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Vimlendu Bhushan Sinha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Ajay Kumar Yadav
- BR Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
| |
Collapse
|
19
|
Malavia N, Kuche K, Ghadi R, Jain S. A bird's eye view of the advanced approaches and strategies for overshadowing triple negative breast cancer. J Control Release 2020; 330:72-100. [PMID: 33321156 DOI: 10.1016/j.jconrel.2020.12.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Triple negative breast cancer (TNBC) is one of the most aggressive form of breast cancer. It is characterized by the absence of estrogen, progesterone and human epidermal growth factor receptors. The main issue with TNBC is that it exhibits poor prognosis, high risk of relapse, short progression-free survival and low overall survival in patients. This is because the conventional therapy used for managing TNBC has issues pertaining to poor bioavailability, lower cellular uptake, increased off-target effects and development of resistance. To overcome such pitfalls, several other approaches are explored. In this context, the present manuscript showcases three of the most widely used approaches which are (i) nanotechnology-based approach; (ii) gene therapy approach and (iii) Phytochemical-based approach. The ultimate focus is to present and explain the insightful reports based on these approaches. Further, the review also expounds on the identified molecular targets and novel targeting ligands which are explored for managing TNBC effectively. Thus, in a nutshell, the review tries to highlight these existing treatment approaches which might inspire for future development of novel therapies with a potential of overshadowing TNBC.
Collapse
Affiliation(s)
- Nilesh Malavia
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Kaushik Kuche
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Rohan Ghadi
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India.
| |
Collapse
|
20
|
Kunjiappan S, Pavadai P, Vellaichamy S, Ram Kumar Pandian S, Ravishankar V, Palanisamy P, Govindaraj S, Srinivasan G, Premanand A, Sankaranarayanan M, Theivendren P. Surface receptor‐mediated targeted drug delivery systems for enhanced cancer treatment: A state‐of‐the‐art review. Drug Dev Res 2020; 82:309-340. [DOI: 10.1002/ddr.21758] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Selvaraj Kunjiappan
- Department of Biotechnology Kalasalingam Academy of Research and Education Krishnankoil Tamilnadu India
| | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy M.S. Ramaiah University of Applied Sciences Bengaluru Karnataka India
| | - Sivakumar Vellaichamy
- Department of Pharmaceutics Arulmigu Kalasalingam College of Pharmacy Krishnankoil Tamilnadu India
| | | | | | - Ponnusamy Palanisamy
- School of Mechanical Engineering Vellore Institute of Technology Vellore Tamilnadu India
| | - Saravanan Govindaraj
- Department of Pharmaceutical Chemistry MNR College of Pharmacy Sangareddy Telangana India
| | - Gowshiki Srinivasan
- Department of Biotechnology Kalasalingam Academy of Research and Education Krishnankoil Tamilnadu India
| | - Adhvitha Premanand
- Department of Biotechnology Kalasalingam Academy of Research and Education Krishnankoil Tamilnadu India
| | | | - Panneerselvam Theivendren
- Department of Pharmaceutical Chemistry Swamy Vivekananda College of Pharmacy Elayampalayam, Namakkal Tamilnadu India
| |
Collapse
|
21
|
Wang W, Liu X, Zheng X, Jin HJ, Li X. Biomineralization: An Opportunity and Challenge of Nanoparticle Drug Delivery Systems for Cancer Therapy. Adv Healthc Mater 2020; 9:e2001117. [PMID: 33043640 DOI: 10.1002/adhm.202001117] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/29/2020] [Indexed: 12/12/2022]
Abstract
Biomineralization is a common process in organisms to produce hard biomaterials by combining inorganic ions with biomacromolecules. Multifunctional nanoplatforms are developed based on the mechanism of biomineralization in many biomedical applications. In the past few years, biomineralization-based nanoparticle drug delivery systems for the cancer treatment have gained a lot of research attention due to the advantages including simple preparation, good biocompatibility, degradability, easy modification, versatility, and targeting. In this review, the research trends of biomineralization-based nanoparticle drug delivery systems and their applications in cancer therapy are summarized. This work aims to promote future researches on cancer therapy based on biomineralization. Rational design of nanoparticle drug delivery systems can overcome the bottleneck in the clinical transformation of nanomaterials. At the same time, biomineralization has also provided new research ideas for cancer treatment, i.e., targeted therapy, which has significantly better performance.
Collapse
Affiliation(s)
- Weicai Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Xiaofan Liu
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Xiangjiang Zheng
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Hyung Jong Jin
- Department of Bioscience and Biotechnology The University of Suwon Hwaseong Gyeonggi‐Do 18323 Republic of Korea
| | - Xuemei Li
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| |
Collapse
|
22
|
Controlled Delivery of BET-PROTACs: In Vitro Evaluation of MZ1-Loaded Polymeric Antibody Conjugated Nanoparticles in Breast Cancer. Pharmaceutics 2020; 12:pharmaceutics12100986. [PMID: 33086530 PMCID: PMC7589709 DOI: 10.3390/pharmaceutics12100986] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/24/2020] [Accepted: 10/15/2020] [Indexed: 12/19/2022] Open
Abstract
Bromo and extraterminal domain (BET) inhibitors-PROteolysis TArgeting Chimera (BETi-PROTAC) is a new family of compounds that induce proteasomal degradation through the ubiquitination of the tagged to BET inhibitors Bromodomain proteins, BRD2 and BRD. The encapsulation and controlled release of BET-PROTACs through their vectorization with antibodies, like trastuzumab, could facilitate their pharmacokinetic and efficacy profile. Antibody conjugated nanoparticles (ACNPs) using PROTACs have not been designed and evaluated. In this pioneer approach, the commercial MZ1 PROTAC was encapsulated into the FDA-approved polymeric nanoparticles. The nanoparticles were conjugated with trastuzumab to guide the delivery of MZ1 to breast tumoral cells that overexpress HER2. These ACNPs were characterized by means of size, polydispersity index, and Z-potential. Morphology of the nanoparticles, along with stability and release studies, completed the characterization. MZ1-loaded ACNPs showed a significant cytotoxic effect maintaining its mechanism of action and improving its therapeutic properties.
Collapse
|
23
|
Juan A, Cimas FJ, Bravo I, Pandiella A, Ocaña A, Alonso-Moreno C. Antibody Conjugation of Nanoparticles as Therapeutics for Breast Cancer Treatment. Int J Mol Sci 2020; 21:E6018. [PMID: 32825618 PMCID: PMC7504566 DOI: 10.3390/ijms21176018] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 02/08/2023] Open
Abstract
Breast cancer is the most common invasive tumor in women and the second leading cause of cancer-related death. Nanomedicine raises high expectations for millions of patients as it can provide better, more efficient, and affordable healthcare, and it has the potential to develop novel therapeutics for the treatment of solid tumors. In this regard, targeted therapies can be encapsulated into nanocarriers, and these nanovehicles are guided to the tumors through conjugation with antibodies-the so-called antibody-conjugated nanoparticles (ACNPs). ACNPs can preserve the chemical structure of drugs, deliver them in a controlled manner, and reduce toxicity. As certain breast cancer subtypes and indications have limited therapeutic options, this field provides hope for the future treatment of patients with difficult to treat breast cancers. In this review, we discuss the application of ACNPs for the treatment of this disease. Given the fact that ACNPs have shown clinical activity in this clinical setting, special emphasis on the role of the nanovehicles and their translation to the clinic is placed on the revision.
Collapse
Affiliation(s)
- Alberto Juan
- Oncología Traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
| | - Francisco J. Cimas
- Oncología Traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Centro Regional de Investigaciones Biomédicas, Unidad Oncología Traslacional, 02071 Albacete, Spain
| | - Iván Bravo
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer-CSIC, IBSAL- Salamanca and CIBERONC, 37007 Salamanca, Spain;
| | - Alberto Ocaña
- Oncología Traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Experimental Therapeutics Unit, Hospital clínico San Carlos, IdISSC and CIBERONC, 28040 Madrid, Spain
| | - Carlos Alonso-Moreno
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| |
Collapse
|
24
|
Johnston MC, Nicoll JA, Redmond KM, Smyth P, Greene MK, McDaid WJ, Chan DKW, Crawford N, Stott KJ, Fox JP, Straubinger NL, Roche S, Clynes M, Straubinger RM, Longley DB, Scott CJ. DR5-targeted, chemotherapeutic drug-loaded nanoparticles induce apoptosis and tumor regression in pancreatic cancer in vivo models. J Control Release 2020; 324:610-619. [PMID: 32504778 PMCID: PMC7429293 DOI: 10.1016/j.jconrel.2020.05.046] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/18/2020] [Accepted: 05/28/2020] [Indexed: 12/31/2022]
Abstract
Pancreatic cancer is usually advanced and drug resistant at diagnosis. A potential therapeutic approach outlined here uses nanoparticle (NP)-based drug carriers, which have unique properties that enhance intra-tumor drug exposure and reduce systemic toxicity of encapsulated drugs. Here we report that patients whose pancreatic cancers express elevated levels of Death Receptor 5 (DR5) and its downstream regulators/effectors FLIP, Caspase-8, and FADD had particularly poor prognoses. To take advantage of elevated expression of this pathway, we designed drug-loaded NPs with a surface-conjugated αDR5 antibody (AMG 655). Binding and clustering of the DR5 is a prerequisite for efficient apoptosis initiation, and the αDR5-NPs were indeed found to activate apoptosis in multiple pancreatic cancer models, whereas the free antibody did not. The extent of apoptosis induced by αDR5-NPs was enhanced by down-regulating FLIP, a key modulator of death receptor-mediated activation of caspase-8. Moreover, the DNA topoisomerase-1 inhibitor camptothecin (CPT) down-regulated FLIP in pancreatic cancer models and enhanced apoptosis induced by αDR5-NPs. CPT-loaded αDR5-NPs significantly increased apoptosis and decreased cell viability in vitro in a caspase-8- and FADD-dependent manner consistent with their expected mechanism-of-action. Importantly, CPT-loaded αDR5-NPs markedly reduced tumor growth rates in vivo in established pancreatic tumor models, inducing regressions in one model. These proof-of-concept studies indicate that αDR5-NPs loaded with agents that downregulate or inhibit FLIP are promising candidate agents for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Michael C Johnston
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Ireland
| | - Julie A Nicoll
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Ireland
| | - Kelly M Redmond
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Ireland
| | - Peter Smyth
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Ireland
| | - Michelle K Greene
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Ireland
| | - William J McDaid
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Ireland
| | - Darren K W Chan
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, United States of America
| | - N Crawford
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Ireland
| | - Katie J Stott
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Ireland
| | - Jennifer P Fox
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Ireland
| | - Ninfa L Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, United States of America
| | - Sandra Roche
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Robert M Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, United States of America; Department of Molecular and Cellular Biophysics and Biochemistry, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States of America; Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States of America
| | - Daniel B Longley
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Ireland.
| | - Christopher J Scott
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Ireland.
| |
Collapse
|
25
|
Szymański W, Gornowicz A, Bielawska A, Bielawski K. The application of nanotechnology in cancer immunotherapy. POSTEP HIG MED DOSW 2020. [DOI: 10.5604/01.3001.0014.1527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Targeted therapy is associated with the use of drugs designed against specific molecular targets. Their mechanism of action is based on the inhibition of specific signaling pathways in processes related to the development of cancer (proliferation, invasion, angiogenesis or metastasis). One of the most important methods of treatment is immunotherapy, which uses monoclonal antibodies. Their mechanism of action is based on inducing programmed cell death by inhibiting specific signal transduction processes. However, immunotherapy has a number of limitations, including side effects that may endanger the patient’s life. To overcome those obstacles immunoconjugates were developed, which combine a monoclonal antibody, or its fragment, with
a drug using a stable linker. Their mechanism of action is based on the monoclonal antibody
binding to a cell membrane receptor, their internalization, the degradation of the linker, and the
release of the drug attached to the antibody, which then activates specific genes or proteins or
induces apoptosis. Immunoconjugates represent a promising alternative for anticancer treatment
used today, but their use is associated with some obstacles. Nanotechnology helps to solve
these problems with a chemotherapeutics delivery system called immunonanoparticles. It uses
chemotherapeutics encapsulated in nanoparticles in combination with monoclonal antibodies
displaying the ability of selective recognition and binding with molecular targets on the surface
of selected cancer cells. This review focuses on presenting the most important solutions used
in targeted therapy, which combine traditional immunotherapy with modern nanotechnology.
Collapse
Affiliation(s)
| | | | - Anna Bielawska
- Zakład Biotechnologii, Uniwersytet Medyczny w Białymstoku
| | - Krzysztof Bielawski
- Zakład Syntezy i Technologii Środków Leczniczych, Uniwersytet Medyczny w Białymstoku
| |
Collapse
|
26
|
Fan Q, Cui X, Guo H, Xu Y, Zhang G, Peng B. Application of rare earth-doped nanoparticles in biological imaging and tumor treatment. J Biomater Appl 2020; 35:237-263. [DOI: 10.1177/0885328220924540] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Rare earth-doped nanoparticles have been widely used in disease diagnosis, drug delivery, tumor therapy, and bioimaging. Among various bioimaging methods, the fluorescence imaging technology based on the rare earth-doped nanoparticles can visually display the cell activity and lesion evolution in living animals, which is a powerful tool in biological technology and has being widely applied in medical and biological fields. Especially in the band of near infrared (700–1700 nm), the emissions show the characteristics of deep penetration due to low absorption, low photon scattering, and low autofluorescence interference. Furthermore, the rare earth-doped nanoparticles can be endowed with the water solubility, biocompatibility, drug-loading ability, and the targeting ability for different tumors by surface functionalization. This confirms its potential in the cancer diagnosis and treatment. In this review, we summarized the recent progress in the application of rare earth-doped nanoparticles in the field of bioimaging and tumor treatment. The luminescent mechanism, properties, and structure design were also discussed.
Collapse
Affiliation(s)
- Qi Fan
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- University of Chinese Academy of Sciences (UCAS), Beijing, PR China
| | - Xiaoxia Cui
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| | - Haitao Guo
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| | - Yantao Xu
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| | - Guangwei Zhang
- Zhejiang Fountain Aptitude Technology Inc., Hangzhou, Zhejiang, PR China
| | - Bo Peng
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| |
Collapse
|
27
|
Targeting strategies for superparamagnetic iron oxide nanoparticles in cancer therapy. Acta Biomater 2020; 102:13-34. [PMID: 31759124 DOI: 10.1016/j.actbio.2019.11.027] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/01/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022]
Abstract
Among various nanoparticles, superparamagnetic iron oxide nanoparticles (SPIONs) have been increasingly studied for their excellent superparamagnetism, magnetic heating properties, and enhanced magnetic resonance imaging (MRI). The conjugation of SPIONs with drugs to obtain delivery nanosystems has several advantages including magnetic targeted functionalization, in vivo imaging, magnetic thermotherapy, and combined delivery of anticancer agents. To further increase the targeting efficiency of drugs through a delivery nanosystem based on SPIONs, additional targeting moieties including transferrin, antibodies, aptamers, hyaluronic acid, folate, and targeting peptides are coated onto the surface of SPIONs. Therefore, this review summarizes the latest progresses in the conjugation of targeting molecules and drug delivery nanosystems based on SPIONs, especially focusing on their performances to develop efficient targeted drug delivery systems for tumor therapy. STATEMENT OF SIGNIFICANCE: Some magnetic nanoparticle-based nanocarriers loaded with drugs were evaluated in patients and did not produce convincing results, leading to termination of clinical development in phase II/III. An alternative strategy for drug delivery systems based on SPIONs is the conjugation of these systems with targeting segments such as transferrin, antibodies, aptamers, hyaluronic acid, folate, and targeting peptides. These targeting moieties can be recognized by specific integrin/receptors that are overexpressed specifically on the tumor cell surface, resulting in minimizing dosage and reducing off-target effects. This review focuses on magnetic nanoparticle-based nonviral drug delivery systems with targeting moieties to deliver anticancer drugs, with an aim to provide suggestions on the development of SPIONs through discussion.
Collapse
|
28
|
McDaid WJ, Greene MK, Johnston MC, Pollheimer E, Smyth P, McLaughlin K, Van Schaeybroeck S, Straubinger RM, Longley DB, Scott CJ. Repurposing of Cetuximab in antibody-directed chemotherapy-loaded nanoparticles in EGFR therapy-resistant pancreatic tumours. NANOSCALE 2019; 11:20261-20273. [PMID: 31626255 PMCID: PMC6861736 DOI: 10.1039/c9nr07257h] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The anti-Epidermal Growth Factor Receptor (EGFR) antibody Cetuximab (CTX) has demonstrated limited anti-cancer efficacy in cells overexpressing EGFR due to activating mutations in RAS in solid tumours, such as pancreatic cancer. The utilisation of antibodies as targeting components of antibody-drug conjugates, such as trastuzumab emtansine (Kadcyla), demonstrates that antibodies may be repurposed to direct therapeutic agents to antibody-resistant cancers. Here we investigated the use of CTX as a targeting agent for camptothecin (CPT)-loaded polymeric nanoparticles (NPs) directed against KRAS mutant CTX-resistant cancer cells. CPT was encapsulated within poly(lactic-co-glycolic acid) (PLGA) NPs using the solvent evaporation method. CTX conjugation improved NP binding and delivery of CPT to CTX-resistant cancer cell lines. CTX successfully targeted CPT-loaded NPs to mutant KRAS PANC-1 tumours in vivo and reduced tumour growth. This study highlights that CTX can be repurposed as a targeting agent against CTX-resistant cancers and that antibody repositioning may be applicable to other antibodies restricted by resistance.
Collapse
Affiliation(s)
- William J McDaid
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Michelle K Greene
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Michael C Johnston
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Ellen Pollheimer
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Peter Smyth
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Kirsty McLaughlin
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | | | - Robert M Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Amherst, NY 14260-1200, USA
| | - Daniel B Longley
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Christopher J Scott
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
29
|
Zirconium-89 radio-nanochemistry and its applications towards the bioimaging of prostate cancer. Inorganica Chim Acta 2019. [DOI: 10.1016/j.ica.2019.119041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
30
|
Dal Corso A, Pignataro L, Belvisi L, Gennari C. Innovative Linker Strategies for Tumor‐Targeted Drug Conjugates. Chemistry 2019; 25:14740-14757. [DOI: 10.1002/chem.201903127] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/15/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Alberto Dal Corso
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| | - Luca Pignataro
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| | - Laura Belvisi
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| | - Cesare Gennari
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| |
Collapse
|
31
|
Farahavar G, Abolmaali SS, Gholijani N, Nejatollahi F. Antibody-guided nanomedicines as novel breakthrough therapeutic, diagnostic and theranostic tools. Biomater Sci 2019; 7:4000-4016. [PMID: 31355391 DOI: 10.1039/c9bm00931k] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recent advances in nanotechnology, such as the development of various types of nanoparticles and hybrid nanomaterials, have revolutionized nanomedicine. The small size, customizable surface, enhanced solubility, and multi-functionality endow the nanoparticles with an ability to interact with complex cellular and biological functions in new ways. Furthermore, these systems can deliver drugs to specific tissues and provide a targeted therapy. For this purpose, different categories of molecules, particularly antibodies, have been used as ligands. Antibody-conjugated nanomaterials can significantly enhance the efficiency of nanomedicines, especially in the field of cancer. This review is focused on three major medical applications of antibody-conjugated nanomaterials, namely, therapeutic, diagnostic and theranostic applications. To provide comprehensive information on the topic and an overview of these hybrid nanomaterials for biomedical applications, a brief summary of nanomaterials and antibodies is given. Moreover, the review has depicted the potential applications of antibody-conjugated nanomaterials in different fields and their capabilities to empower nanomedicine, particularly in relation to the treatment and detection of malignancies.
Collapse
Affiliation(s)
- Ghazal Farahavar
- Pharmaceutical Nanotechnology Department, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Samira Sadat Abolmaali
- Pharmaceutical Nanotechnology Department and Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Nasser Gholijani
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Foroogh Nejatollahi
- Shiraz HIV/AIDS research center, Institute of health, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
32
|
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: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
33
|
Belousov AV, Morozov VN, Krusanov GA, Kolyvanova MA, Shtil AA. The Effect of Gold Nanoparticle Surface Modification with Polyethylene Glycol on the Absorbed Dose Distribution upon Irradiation with 137Cs and 60Co Photons. Biophysics (Nagoya-shi) 2019. [DOI: 10.1134/s0006350919010032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
34
|
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
|
35
|
Bae CS, Ahn T. Diacylglycerol in Cationic Nanoparticles Stimulates Oxidative Stress-Mediated Death of Cancer Cells. Lipids 2019; 53:1059-1067. [DOI: 10.1002/lipd.12124] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/20/2018] [Accepted: 12/24/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Chun-Sik Bae
- College of Veterinary Medicine; Chonnam National University; 77 Yongbong-ro, Buk-gu, Gwangju 61186 Republic of Korea
| | - Taeho Ahn
- College of Veterinary Medicine; Chonnam National University; 77 Yongbong-ro, Buk-gu, Gwangju 61186 Republic of Korea
| |
Collapse
|
36
|
Johnston MC, Scott CJ. Antibody conjugated nanoparticles as a novel form of antibody drug conjugate chemotherapy. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:63-69. [PMID: 30553522 DOI: 10.1016/j.ddtec.2018.10.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 06/09/2023]
Abstract
Antibody conjugated nanoparticles (ACNPs) represent a novel strategy for the development of therapies exploiting antibodies to augment the delivery of chemotherapy payloads. Following in the footsteps of the success of antibody drug conjugates (ADCs), ACNPs are only now reaching clinical evaluation. In this review we discuss the success of ADCs and explore the opportunities ACNPs offer, such as broad chemotherapy payload selection, high drug to antibody ratios and the ability to finely tailor drug release in comparison to ADCs. The ability of ACNPs to elicit increased avidity due to multivalent effects and the potential to use these modular platforms in immunotherapeutic approaches is also explored. Through addressing challenges that still remain in bringing these complex formulations to the clinic, ACNPs hold obvious potential for the treatment of a wide range of cancers and other diseases where selective targeting of drug agents is essential.
Collapse
Affiliation(s)
- Michael C Johnston
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, United Kingdom
| | - Christopher J Scott
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, United Kingdom.
| |
Collapse
|
37
|
Light-induced mechanisms for nanocarrier's cargo release. Colloids Surf B Biointerfaces 2018; 173:825-832. [PMID: 30551298 DOI: 10.1016/j.colsurfb.2018.10.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 01/18/2023]
Abstract
Nanomaterials have been the focus of attention in several fields, including biomedicine, electronics, or catalysis, mainly due to the novel properties of the materials at the nanoscale. In the field of diagnosis, nanomaterials have been contemplated as an opportunity to improve sensitivity and time of response, therefore, facilitating early treatment and monitoring of the disease. For therapeutic applications, new drug delivery nanosystems aiming to provide enhanced efficiency have been proposed often addressing selective or controlled delivery of therapeutic agents to particular cells to maximize treatment efficacy minimizing adverse effects. The therapeutic agents can be dissolved, adsorbed, entrapped, encapsulated or attached on the surface or inside the nanocarriers. Given the context of the different generations of nanocarriers and their wide range of applications, the present article aims to discuss the nature of external stimuli which will trigger the controlled release of different biomolecules. For each class, a brief description of the physical principle, basic concepts, as well as some examples, are reported. A final discussion focused on the real implications and needs for optimal drug delivery system is presented, altogether with some considerations and prospects in the trends that diagnostics applications could follow in the next years.
Collapse
|
38
|
Noor A, Walser G, Wesseling M, Giron P, Laffra AM, Haddouchi F, De Grève J, Kronenberger P. Production of a mono-biotinylated EGFR nanobody in the E. coli periplasm using the pET22b vector. BMC Res Notes 2018; 11:751. [PMID: 30348204 PMCID: PMC6196415 DOI: 10.1186/s13104-018-3852-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/12/2018] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVE Our aim was to produce a mono-biotinylated single domain antibody ('nanobody') specific for the epidermal growth factor receptor (EGFR), which is overexpressed in many cancer cells. The binding of the nanobody and its function are tested in cancer cells. The construct could be used to carry variable therapeutic or diagnostic load using biotin-streptavidin bridging. RESULTS The EGFR-specific 7D12 nanobody was genetically fused to an IgA hinge linker and to a C-terminal biotin ligase acceptor sequence, allowing mono-biotinylation in E. coli. Expression was in strain BL21-DE3 from a T7 RNA polymerase driven pET22b vector. The biotinylated nanobody, isolated from the periplasm, was purified using streptavidin-mutein affinity chromatography. Final yields were up to 5 mg/l of cell culture. We showed that the construct could bind to EGFR expressing A431 epidermoid carcinoma cells, and to transiently transformed EGFR overexpressing HEK293T cells and not to EGFR negative control cells. The specificity for the EGFR was further demonstrated by immunoprecipitation. To test the functionality, PC9 non-small cell lung cancer cells were treated with mono-biotinylated nanobody or with streptavidin-coupled tetravalent nanobodies. Both were able to block mutant EGFR phosphorylation and slow down growth of PC9 cells. Tetravalent nanobodies were able to downregulate AKT phosphorylation.
Collapse
Affiliation(s)
- Alfiah Noor
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, Laarbeeklaan 109, 1090, Brussels, Belgium.
| | - Gudrun Walser
- Laboratory of Biotechnology, Erasmushogeschool Brussel, Laarbeeklaan 121, 1090, Brussels, Belgium
| | - Matthijs Wesseling
- Laboratory of Biotechnology, Erasmushogeschool Brussel, Laarbeeklaan 121, 1090, Brussels, Belgium
| | - Philippe Giron
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, Laarbeeklaan 109, 1090, Brussels, Belgium
| | - Albert-Menno Laffra
- Laboratory of Biotechnology, Erasmushogeschool Brussel, Laarbeeklaan 121, 1090, Brussels, Belgium
| | - Fatima Haddouchi
- Laboratory of Biotechnology, Erasmushogeschool Brussel, Laarbeeklaan 121, 1090, Brussels, Belgium
| | - Jacques De Grève
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, Laarbeeklaan 109, 1090, Brussels, Belgium
| | - Peter Kronenberger
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, Laarbeeklaan 109, 1090, Brussels, Belgium. .,Laboratory of Biotechnology, Erasmushogeschool Brussel, Laarbeeklaan 121, 1090, Brussels, Belgium.
| |
Collapse
|
39
|
Tallian C, Herrero-Rollett A, Stadler K, Vielnascher R, Wieland K, Weihs AM, Pellis A, Teuschl AH, Lendl B, Amenitsch H, Guebitz GM. Structural insights into pH-responsive drug release of self-assembling human serum albumin-silk fibroin nanocapsules. Eur J Pharm Biopharm 2018; 133:176-187. [PMID: 30291964 DOI: 10.1016/j.ejpb.2018.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/05/2018] [Accepted: 10/01/2018] [Indexed: 12/22/2022]
Abstract
Inflammation processes are associated with significant decreases in tissue or lysosomal pH from 7.4 to 4, a fact that argues for the application of pH-responsive drug delivery systems. However, for their design and optimization a full understanding of the release mechanism is crucial. In this study we investigated the pH-depending drug release mechanism and the influence of silk fibroin (SF) concentration and SF degradation degree of human serum albumin (HSA)-SF nanocapsules. Sonochemically produced nanocapsules were investigated regarding particle size, colloidal stability, protein encapsulation, thermal stability and drug loading properties. Particles of the monodisperse phase showed average hydrodynamic radii between 438 and 888 nm as measured by DLS and AFM and a zeta potential of -11.12 ± 3.27 mV. Together with DSC results this indicated the successful production of stable nanocapsules. ATR-FTIR analysis demonstrated that SF had a positive effect on particle formation and stability due to induced beta-sheet formation and enhanced crosslinking. The pH-responsive release was found to depend on the SF concentration. In in-vitro release studies, HSA-SF nanocapsules composed of 50% SF showed an increased pH-responsive release for all tested model substances (Rhodamine B, Crystal Violet and Evans Blue) and methotrexate at the lowered pH of 4.5 to pH 5.4, while HSA capsules without SF did not show any pH-responsive drug release. Mechanistic studies using confocal laser scanning microscopy (CLSM) and small angle X-ray scattering (SAXS) analyses showed that increases in particle porosity and decreases in particle densities are directly linked to pH-responsive release properties. Therefore, the pH-responsive release mechanism was identified as diffusion controlled in a novel and unique approach by linking scattering results with in-vitro studies. Finally, cytotoxicity studies using the human monocytic THP-1 cell line indicated non-toxic behavior of the drug loaded nanocapsules when applied in a concentration of 62.5 µg mL-1. Based on the obtained release properties of HSA-SF nanocapsules formulations and the results of in-vitro MTT assays, formulations containing 50% SF showed the highest requirements arguing for future in vivo experiments and application in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Claudia Tallian
- Institute of Environmental Biotechnology, Department of Agrobiotechnology (IFA-Tulln), University of Natural Resources and Life Sciences, Vienna (BOKU), Konrad-Lorenz-Straße 20, 3430 Tulln an der Donau, Austria
| | - Alexandra Herrero-Rollett
- Institute of Environmental Biotechnology, Department of Agrobiotechnology (IFA-Tulln), University of Natural Resources and Life Sciences, Vienna (BOKU), Konrad-Lorenz-Straße 20, 3430 Tulln an der Donau, Austria.
| | - Karina Stadler
- Institute of Environmental Biotechnology, Department of Agrobiotechnology (IFA-Tulln), University of Natural Resources and Life Sciences, Vienna (BOKU), Konrad-Lorenz-Straße 20, 3430 Tulln an der Donau, Austria
| | - Robert Vielnascher
- Institute of Environmental Biotechnology, Department of Agrobiotechnology (IFA-Tulln), University of Natural Resources and Life Sciences, Vienna (BOKU), Konrad-Lorenz-Straße 20, 3430 Tulln an der Donau, Austria; ACIB - Austrian Centre of Industrial Biotechnology, Konrad-Lorenz-Straße 20, 3430 Tulln an der Donau, Austria
| | - Karin Wieland
- Institute of Chemical Technologies and Analytics, Division of Analytical Chemistry, Vienna University of Technology, Getreidemarkt 9/164 AC, 1060 Vienna, Austria
| | - Anna M Weihs
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Alessandro Pellis
- Institute of Environmental Biotechnology, Department of Agrobiotechnology (IFA-Tulln), University of Natural Resources and Life Sciences, Vienna (BOKU), Konrad-Lorenz-Straße 20, 3430 Tulln an der Donau, Austria; Department of Chemistry, University of York, Heslington, York YO10 5DD, United Kingdom
| | - Andreas H Teuschl
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Bernhard Lendl
- Institute of Chemical Technologies and Analytics, Division of Analytical Chemistry, Vienna University of Technology, Getreidemarkt 9/164 AC, 1060 Vienna, Austria
| | - Heinz Amenitsch
- Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse 9/IV, 8010 Graz, Austria
| | - Georg M Guebitz
- Institute of Environmental Biotechnology, Department of Agrobiotechnology (IFA-Tulln), University of Natural Resources and Life Sciences, Vienna (BOKU), Konrad-Lorenz-Straße 20, 3430 Tulln an der Donau, Austria; ACIB - Austrian Centre of Industrial Biotechnology, Konrad-Lorenz-Straße 20, 3430 Tulln an der Donau, Austria.
| |
Collapse
|
40
|
Gibson TJ, Smyth P, McDaid WJ, Lavery D, Thom J, Cotton G, Scott CJ, Themistou E. Single-Domain Antibody-Functionalized pH-Responsive Amphiphilic Block Copolymer Nanoparticles for Epidermal Growth Factor Receptor Targeted Cancer Therapy. ACS Macro Lett 2018; 7:1010-1015. [PMID: 35650954 DOI: 10.1021/acsmacrolett.8b00461] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Biocompatible antibody-nanoparticle conjugates have attracted interest as anticancer agents due to their potential to selectively target therapeutic agents at disease sites. However, new formulation and conjugation approaches are urgently needed to improve their uniformity for clinical applications. Here, a pH-responsive benzaldehyde-functionalized poly[oligo(ethylene glycol) methacrylate-st-para-formyl phenyl methacrylate]-b-poly[2-(diisopropyl)aminoethyl methacrylate] [P(OEGMA-st-pFPMA)-b-PDPA] block copolymer, prepared by reversible addition-fragmentation chain transfer polymerization, produced PEGylated nanoparticles (pH ∼ 7.4) by a single emulsion-solvent evaporation formulation approach. Efficient site-specific attachment of an aminooxy-functionalized anti-EGFR single-domain antibody (sdAb) on these benzaldehyde-decorated nanoparticles is achieved by oxime bond formation. These nanoconjugates can specifically bind EGFR (modified ELISA) and have enhanced uptake over nonfunctionalized controls in EGFR-positive HeLa cells. Encapsulation of rhodamine 6G dye and its dispersion upon cellular uptake, consistent with nanoparticle stability loss at pH < 5.7, prove their ability to facilitate triggered release in endosomal compartments and highlight their potential for use as next-generation antibody-drug nanoconjugates for therapeutic drug delivery.
Collapse
Affiliation(s)
- Thomas J. Gibson
- School of Chemistry and Chemical Engineering, Queen’s University Belfast, Belfast BT9 5AG, U.K
| | - Peter Smyth
- Centre for Cancer Research & Cell Biology, Queen’s University Belfast, Belfast BT9 7AE, U.K
| | - William J. McDaid
- Centre for Cancer Research & Cell Biology, Queen’s University Belfast, Belfast BT9 7AE, U.K
| | - Daniel Lavery
- School of Chemistry and Chemical Engineering, Queen’s University Belfast, Belfast BT9 5AG, U.K
| | - Jennifer Thom
- Almac Discovery, The Fleming Building, Edinburgh Technopole, Edinburgh EH26 0BE, U.K
| | - Graham Cotton
- Almac Discovery, The Fleming Building, Edinburgh Technopole, Edinburgh EH26 0BE, U.K
| | - Christopher J. Scott
- Centre for Cancer Research & Cell Biology, Queen’s University Belfast, Belfast BT9 7AE, U.K
| | - Efrosyni Themistou
- School of Chemistry and Chemical Engineering, Queen’s University Belfast, Belfast BT9 5AG, U.K
| |
Collapse
|
41
|
Belousov AV, Morozov VN, Krusanov GА, Kolyvanova MA, Chernyaev AP, Shtil AA. Spectra of secondary particles generated upon virtual irradiation of gold nanosensitizers: implications for surface modification. Biomed Phys Eng Express 2018. [DOI: 10.1088/2057-1976/aac73d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
42
|
Namdee K, Khongkow M, Boonthod S, Boonrungsiman S, Jarussophon S, Pongwan P, Yata T, Saengkrit N. Cell-based assay for characterizing cell adhesion properties of active targeted nanoparticles under static and flow condition using an integrated flow chamber. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
43
|
Spicer CD, Jumeaux C, Gupta B, Stevens MM. Peptide and protein nanoparticle conjugates: versatile platforms for biomedical applications. Chem Soc Rev 2018; 47:3574-3620. [PMID: 29479622 PMCID: PMC6386136 DOI: 10.1039/c7cs00877e] [Citation(s) in RCA: 295] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Peptide- and protein-nanoparticle conjugates have emerged as powerful tools for biomedical applications, enabling the treatment, diagnosis, and prevention of disease. In this review, we focus on the key roles played by peptides and proteins in improving, controlling, and defining the performance of nanotechnologies. Within this framework, we provide a comprehensive overview of the key sequences and structures utilised to provide biological and physical stability to nano-constructs, direct particles to their target and influence their cellular and tissue distribution, induce and control biological responses, and form polypeptide self-assembled nanoparticles. In doing so, we highlight the great advances made by the field, as well as the challenges still faced in achieving the clinical translation of peptide- and protein-functionalised nano-drug delivery vehicles, imaging species, and active therapeutics.
Collapse
Affiliation(s)
- Christopher D Spicer
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, Stockholm, Sweden.
| | | | | | | |
Collapse
|
44
|
Norouzi H, Khoshgard K, Akbarzadeh F. In vitro outlook of gold nanoparticles in photo-thermal therapy: a literature review. Lasers Med Sci 2018; 33:917-926. [PMID: 29492712 DOI: 10.1007/s10103-018-2467-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 02/08/2018] [Indexed: 12/31/2022]
Abstract
Hyperthermia is an anti-cancer treatment in which the temperature of the malignant tumor is increased more than other adjacent normal tissues. Microwave, ultrasound, laser, and radiofrequency sources have been used for hyperthermia of cancerous tissues. In the past decade, near-infrared (NIR) laser for cancer therapy, known as photo-thermal therapy (PTT), was expanded in which the photo-sensitizer agent converts the light photon energy to heat. The heat following PTT can destroy cancer cells. There are some photo-sensitizer agents which have been used for PTT; however, owing to recent advances in nanotechnology, noble metal nanoparticles like gold (Au) nanoparticles (GNPs) have been used successfully in PTT. GNPs have some desirable specifications, including simple and controlled synthesis, small size, high level of biocompatibility, and surface plasmon resonance (SPR). The SPR effect of the GNPs increases the radiative properties like absorption and scattering; therefore, they can be used in PTT. In this article, we reviewed recent in vitro studies of PTT using GNPs in literature. At first, we focus on the physical properties of GNPs, their interaction with infrared radiation, and physical parameters governing the interaction of infrared radiation with the GNPs. Then, we review the passive and active targeting of GNPs using the different coating to induce the thermal damage in cancer cells using low-level laser PPT. The GNPs' cellular internalization into cancer cells is a challenge which is consequently considered. In this review, we also summarize the results of synergistic cancer therapy studies on the combination of radiation therapy as a routine cancer treatment and PTT: in which significant improvement occurs in treatment efficacy.
Collapse
Affiliation(s)
- Hasan Norouzi
- Student Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Karim Khoshgard
- Department of Medical Physics, School of Medicine, Kermanshah University of Medical Sciences, Sorkheh-Lizhe Blvd, P.O. Box: 1568, Kermanshah, Iran.
| | - Fatemeh Akbarzadeh
- Student Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
45
|
Carradori D, Balducci C, Re F, Brambilla D, Le Droumaguet B, Flores O, Gaudin A, Mura S, Forloni G, Ordoñez-Gutierrez L, Wandosell F, Masserini M, Couvreur P, Nicolas J, Andrieux K. Antibody-functionalized polymer nanoparticle leading to memory recovery in Alzheimer's disease-like transgenic mouse model. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 14:609-618. [PMID: 29248676 DOI: 10.1016/j.nano.2017.12.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/18/2017] [Accepted: 12/04/2017] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder related, in part, to the accumulation of amyloid-β peptide (Aβ) and especially the Aβ peptide 1-42 (Aβ1-42). The aim of this study was to design nanocarriers able to: (i) interact with the Aβ1-42 in the blood and promote its elimination through the "sink effect" and (ii) correct the memory defect observed in AD-like transgenic mice. To do so, biodegradable, PEGylated nanoparticles were surface-functionalized with an antibody directed against Aβ1-42. Treatment of AD-like transgenic mice with anti-Aβ1-42-functionalized nanoparticles led to: (i) complete correction of the memory defect; (ii) significant reduction of the Aβ soluble peptide and its oligomer level in the brain and (iii) significant increase of the Aβ levels in plasma. This study represents the first example of Aβ1-42 monoclonal antibody-decorated nanoparticle-based therapy against AD leading to complete correction of the memory defect in an experimental model of AD.
Collapse
Affiliation(s)
- Dario Carradori
- Institut Galien Paris Sud, CNRS UMR 8612, Univ Paris-Sud, Univ. Paris Saclay, Châtenay-Malabry, France
| | | | - Francesca Re
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| | - Davide Brambilla
- Institut Galien Paris Sud, CNRS UMR 8612, Univ Paris-Sud, Univ. Paris Saclay, Châtenay-Malabry, France
| | - Benjamin Le Droumaguet
- Université Paris-Est, Institut de Chimie et des Matériaux Paris-Est (ICMPE), UMR 7182 CNRS-UPEC, 2 rue Henri Dunant, 94320, Thiais, France
| | - Orfeu Flores
- Stab Vida, Madan Parque, Rua dos Inventores, Caparica, Portugal
| | - Alice Gaudin
- Institut Galien Paris Sud, CNRS UMR 8612, Univ Paris-Sud, Univ. Paris Saclay, Châtenay-Malabry, France
| | - Simona Mura
- Institut Galien Paris Sud, CNRS UMR 8612, Univ Paris-Sud, Univ. Paris Saclay, Châtenay-Malabry, France
| | | | | | - Francisco Wandosell
- Centro de Biología Molecular Severo Ochoa CSIC-UAM & CIBERNED, Madrid, Spain
| | - Massimo Masserini
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Patrick Couvreur
- Institut Galien Paris Sud, CNRS UMR 8612, Univ Paris-Sud, Univ. Paris Saclay, Châtenay-Malabry, France
| | - Julien Nicolas
- Institut Galien Paris Sud, CNRS UMR 8612, Univ Paris-Sud, Univ. Paris Saclay, Châtenay-Malabry, France.
| | - Karine Andrieux
- Faculté de Pharmacie de Paris, UTCBS, CNRS UMR 8258, Inserm U1022, Univ. Paris Descartes, Univ. Sorbonne Paris Cité, Paris, France
| |
Collapse
|
46
|
PEGylated polydopamine-coated magnetic nanoparticles for combined targeted chemotherapy and photothermal ablation of tumour cells. Colloids Surf B Biointerfaces 2017; 160:11-21. [DOI: 10.1016/j.colsurfb.2017.09.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 08/25/2017] [Accepted: 09/05/2017] [Indexed: 11/23/2022]
|
47
|
Li D, Chen X, Wang H, Liu J, Zheng M, Fu Y, Yu Y, Zhi J. Cetuximab-conjugated nanodiamonds drug delivery system for enhanced targeting therapy and 3D Raman imaging. JOURNAL OF BIOPHOTONICS 2017; 10:1636-1646. [PMID: 28635183 DOI: 10.1002/jbio.201700011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/31/2017] [Accepted: 05/02/2017] [Indexed: 05/20/2023]
Abstract
In this study, a multicomponent nanodiamonds (NDs)-based targeting drug delivery system, cetuximab-NDs-cisplatin bioconjugate, combining both specific targeting and enhanced therapeutic efficacy capabilities, is developed and characterized. The specific targeting ability of cetuximab-NDs-cisplatin system on human liver hepatocellular carcinoma (HepG2) cells is evaluated through epidermal growth factor receptor (EGFR) blocking experiments, since EGFR is over-expressed on HepG2 cell membrane. Besides, cytotoxic evaluation confirms that cetuximab-NDs-cisplatin system could significantly inhibit the growth of HepG2 cells, and the therapeutic activity of this system is proven to be better than that of both nonspecific NDs-cisplatin conjugate and specific EGF-NDs-cisplatin conjugate. Furthermore, a 3-dimensional (3D) Raman imaging technique is utilized to visualize the targeting efficacy and enhanced internalization of cetuximab-NDs-cisplatin system in HepG2 cells, using the NDs existing in the bioconjugate as Raman probes, based on the characteristic Raman signal of NDs at 1332 cm-1 . These advantageous properties of cetuximab-NDs-cisplatin system propose a prospective imaging and treatment tool for further diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Dandan Li
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 100190, PR China
- University of Chinese Academy of Sciences, 100049, PR China
| | - Xin Chen
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 100190, PR China
- University of Chinese Academy of Sciences, 100049, PR China
- School of Pharmaceutical Sciences, Peking University, 100191, PR China
| | - Hong Wang
- School of Pharmaceutical Sciences, Peking University, 100191, PR China
| | - Jie Liu
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 100190, PR China
| | - Meiling Zheng
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 100190, PR China
| | - Yang Fu
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 100190, PR China
- University of Chinese Academy of Sciences, 100049, PR China
| | - Yuan Yu
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 100190, PR China
| | - Jinfang Zhi
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 100190, PR China
| |
Collapse
|
48
|
Hu Y, Liu C, Muyldermans S. Nanobody-Based Delivery Systems for Diagnosis and Targeted Tumor Therapy. Front Immunol 2017; 8:1442. [PMID: 29163515 PMCID: PMC5673844 DOI: 10.3389/fimmu.2017.01442] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 10/17/2017] [Indexed: 01/01/2023] Open
Abstract
The development of innovative targeted therapeutic approaches are expected to surpass the efficacy of current forms of treatments and cause less damage to healthy cells surrounding the tumor site. Since the first development of targeting agents from hybridoma’s, monoclonal antibodies (mAbs) have been employed to inhibit tumor growth and proliferation directly or to deliver effector molecules to tumor cells. However, the full potential of such a delivery strategy is hampered by the size of mAbs, which will obstruct the targeted delivery system to access the tumor tissue. By serendipity, a new kind of functional homodimeric antibody format was discovered in camelidae, known as heavy-chain antibodies (HCAbs). The cloning of the variable domain of HCAbs produces an attractive minimal-sized alternative for mAbs, referred to as VHH or nanobodies (Nbs). Apart from their dimensions in the single digit nanometer range, the unique characteristics of Nbs combine a high stability and solubility, low immunogenicity and excellent affinity and specificity against all possible targets including tumor markers. This stimulated the development of tumor-targeted therapeutic strategies. Some autonomous Nbs have been shown to act as antagonistic drugs, but more importantly, the targeting capacity of Nbs has been exploited to create drug delivery systems. Obviously, Nb-based targeted cancer therapy is mainly focused toward extracellular tumor markers, since the membrane barrier prevents antibodies to reach the most promising intracellular tumor markers. Potential strategies, such as lentiviral vectors and bacterial type 3 secretion system, are proposed to deliver target-specific Nbs into tumor cells and to block tumor markers intracellularly. Simultaneously, Nbs have also been employed for in vivo molecular imaging to diagnose diseased tissues and to monitor the treatment effects. Here, we review the state of the art and focus on recent developments with Nbs as targeting moieties for drug delivery systems in cancer therapy and cancer imaging.
Collapse
Affiliation(s)
- Yaozhong Hu
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium.,State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Changxiao Liu
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China.,State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
49
|
Khutale GV, Casey A. Synthesis and characterization of a multifunctional gold-doxorubicin nanoparticle system for pH triggered intracellular anticancer drug release. Eur J Pharm Biopharm 2017; 119:372-380. [DOI: 10.1016/j.ejpb.2017.07.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/27/2017] [Accepted: 07/18/2017] [Indexed: 12/31/2022]
|
50
|
Malaspina DC, Longo G, Szleifer I. Behavior of ligand binding assays with crowded surfaces: Molecular model of antigen capture by antibody-conjugated nanoparticles. PLoS One 2017; 12:e0185518. [PMID: 28957393 PMCID: PMC5619776 DOI: 10.1371/journal.pone.0185518] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/14/2017] [Indexed: 12/20/2022] Open
Abstract
Ligand-receptor binding is of utmost importance in several biologically related disciplines. Ligand binding assays (LBA) use the high specificity and high affinity of ligands to detect, target or measure a specific receptors. One particular example of ligand binding assays are Antibody conjugated Nanoparticles (AcNPs), edge-cutting technologies that are present in several novel biomedical approaches for imaging, detection and treatment of diseases. However, the nano-confinement in AcNPs and LBA nanostructures introduces extra complexity in the analysis of ligand-receptor equilibriums. Because antibodies are large voluminous ligands, the effective affinity in AcNPs is often determined by antibody orientation and surface coverage. Moreover, antibodies have two binding sites introducing an extra ligand-receptor binding equilibrium. As consequence of all this, experimental or theoretical studies providing a guidelines for the prediction of the binding behavior in AcNPs are scarce. In this work, we present a set of theoretical calculations to shed light into the complex binding behavior of AcNPs and its implications in biomedical applications. To investigate the ligand-receptor binding on AcNPs, we have used a molecular theory that predicts the probability of different molecular conformations of the system depending on the local environment. We have considered two different pathways for designing these devices: covalently conjugated antibodies and streptavidin-biotin conjugated antibodies. We also explore the effects of surface coverage, bulk concentrations, nanoparticle size and antibody-antigen affinity. Overall, this work offers a series of theoretical predictions that can be used as a guide in the design of antibody conjugated nanoparticles for different applications.
Collapse
Affiliation(s)
- David C. Malaspina
- Biomedical Engineering Department, Northwestern University, Evanston, Illinois, United States of America
| | - Gabriel Longo
- Instituto de Investigaciones Fisicoquímicas, Teóricas y Aplicadas (INIFTA), UNLP, CONICET, La Plata, Argentina
| | - Igal Szleifer
- Biomedical Engineering Department, Northwestern University, Evanston, Illinois, United States of America
- Chemistry Department and Chemistry of Life Processes Institute, Evanston, Illinois, United States of America
- * E-mail:
| |
Collapse
|