1
|
Tan KF, Chia LY, Maki MAA, Cheah SC, In LLA, Kumar PV. Gold nanocomposites in colorectal cancer therapy: characterization, selective cytotoxicity, and migration inhibition. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03839-z. [PMID: 39878813 DOI: 10.1007/s00210-025-03839-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025]
Abstract
The third most prevalent type of cancer in the world, colorectal cancer, poses a significant treatment challenge due to the nonspecific distribution, low efficacy, and high systemic toxicity associated with chemotherapy. To overcome these limitations, a targeted drug delivery system with a high cytotoxicity against cancer cells while maintaining a minimal systemic side effects represents a promising therapeutic approach. Therefore, the aim of this study was to develop an efficient gold nanocarrier for the targeted delivery of the anticancer agent everolimus to Caco-2 cells. A novel gold nanocomposite (EV-β-CD-HA-Chi-AuNCs) functionalized with a targeting ligand (hyaluronic acid), a permeation enhancement excipient (chitosan), and an anticancer inclusive compound consisting of beta-cyclodextrin and everolimus was proposed and prepared via Turkevich method. Characterization was performed with a UV spectrometer, FTIR, Zetasizer, and HRTEM. Its drug release profile was also evaluated in media with three different pH values. Cytotoxicity and biocompatibility studies were performed on a colorectal cancer cell line (Caco-2) and a normal fibroblast line (MRC-5), respectively, via xCELLigence real-time cellular analysis (RTCA) technology. The inhibitory effect on migration was also further tested via the xCELLigence RTCA technique and a scratch assay. Characterization studies revealed the successful formation of EV-β-CD-HA-Chi-AuNCs with a size and charge which are suitable for the use as targeted drug delivery carrier. In the cytotoxic study, the EV-β-CD-HA-Chi-AuNCs showed a lower IC50 (16 ± 1 µg/ml) than the pure drug (25 ± 3 µg/ml) toward a colorectal cell line (Caco-2). In the biocompatibility study, the EV-β-CD-HA-Chi-AuNCs have minimal toxicity, while the pure drug has severe toxicity toward healthy fibroblasts (MRC-5) despite its low concentration. In the cell migration study, the EV-β-CD-HA-Chi-AuNCs also showed a greater inhibitory effect than the pure drug. Compared with the pure drug, the EV-β-CD-HA-Chi-AuNCs exhibit an excellent selective cytotoxicity between cancerous colorectal Caco-2 cells and healthy MRC-5 cells, making it a potential carrier to carry the drug to the cancerous site while maintaining its low toxicity to the surrounding environment. In addition, an increase in the cytotoxic activity of the EV-β-CD-HA-Chi-AuNCs toward cancerous colorectal Caco-2 cells was also observed, which can potentially improve the treatment of colorectal cancer.
Collapse
Grants
- REIG-FPS-2023-042 Research Excellence and Innovation Grant under Centre of Excellence in Research, Value Innovation and Entrepreneurship (CERVIE), UCSI University, Malaysia
- REIG-FPS-2023-042 Research Excellence and Innovation Grant under Centre of Excellence in Research, Value Innovation and Entrepreneurship (CERVIE), UCSI University, Malaysia
- REIG-FPS-2023-042 Research Excellence and Innovation Grant under Centre of Excellence in Research, Value Innovation and Entrepreneurship (CERVIE), UCSI University, Malaysia
- FRGS/1/2021/SKK0/UCSI/02/5 Ministry of Higher Education (MOHE), Malaysia
- FRGS/1/2021/SKK0/UCSI/02/5 Ministry of Higher Education (MOHE), Malaysia
- FRGS/1/2021/SKK0/UCSI/02/5 Ministry of Higher Education (MOHE), Malaysia
Collapse
Affiliation(s)
- Kin Fai Tan
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Le Yi Chia
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Marwan Abdelmahmoud Abdelkarim Maki
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Shiau-Chuen Cheah
- Faculty of Medicine and Health Sciences, UCSI University, Bandar Springhill, Port Dickson, Negeri Sembilan, 71010, Malaysia
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia
| | - Palanirajan Vijayaraj Kumar
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur, 56000, Malaysia.
| |
Collapse
|
2
|
Muhammad FA, Altalbawy FMA, Mandaliya V, Saraswat SK, Rekha MM, Aulakh D, Chahar M, Mahdi MS, Jaber MA, Alhadrawi M. Targeting breast tumor extracellular matrix and stroma utilizing nanoparticles. Clin Transl Oncol 2024:10.1007/s12094-024-03793-x. [PMID: 39692807 DOI: 10.1007/s12094-024-03793-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024]
Abstract
Breast cancer is a complicated malignancy and is known as the most common cancer in women. Considerable experiments have been devoted to explore the basic impacts of the tumor stroma, particularly the extracellular matrix (ECM) and stromal components, on tumor growth and resistance to treatment. ECM is made up of an intricate system of proteins, glycosaminoglycans, and proteoglycans, and maintains structural support and controls key signaling pathways involved in breast tumors. ECM can block different drugs such as chemotherapy and immunotherapy drugs from entering the tumor stroma. Furthermore, the stromal elements, such as cancer-associated fibroblasts (CAFs), immune cells, and blood vessels, have crucial impacts on tumor development and therapeutic resistance. Recently, promising outcomes have been achieved in using nanotechnology for delivering drugs to tumor stroma and crossing ECM in breast malignancies. Nanoparticles have various benefits for targeting the breast tumor stroma, such as improved permeability and retention, extended circulation time, and the ability to actively target the area. This review covers the latest developments in nanoparticle therapies that focus on breast tumor ECM and stroma. We will explore different approaches using nanoparticles to target the delivery of anticancer drugs like chemotherapy, small molecule drugs, various antitumor products, and other specific synthetic therapeutic agents to the breast tumor stroma. Furthermore, we will investigate the utilization of nanoparticles in altering the stromal elements, such as reprogramming CAFs and immune cells, and also remodeling ECM.
Collapse
Affiliation(s)
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, 12613, Egypt.
| | - Viralkumar Mandaliya
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | | | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Damanjeet Aulakh
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology Chitkara University, Rajpura, Punjab, 140401, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering and Technology, NIMS University Rajasthan, Jaipur, India
| | | | | | - Merwa Alhadrawi
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
3
|
Tian S, Chen M. Global research progress of nanomedicine and colorectal cancer: a bibliometrics and visualization analysis. Front Oncol 2024; 14:1460201. [PMID: 39711965 PMCID: PMC11660184 DOI: 10.3389/fonc.2024.1460201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/04/2024] [Indexed: 12/24/2024] Open
Abstract
Background Surgery and chemoradiotherapy are the main clinical treatment methods for colorectal cancer (CRC), but the prognosis is poor. The emergence of nanomedicine brings bright light to the treatment of CRC. However, there has not been a comprehensive and systematic analysis of CRC and nanomedicine by bibliometrics. Methods We searched the Web of Science Core Collection database (WOSCC) for relevant literature published from 2011 to 2024. We used VOSviewer and Citespace to analyze countries, institutions, authors, keywords, highly cited references, and co-cited references. Results 3105 pieces of literatures were included in the research analysis, and PEOPLES R CHINA and the USA took the leading position in the number of papers published and had academic influence. The Chinese Academy of Sciences posted the most papers. The most prolific scholar was Abnous Khalil. The level of economic development is inversely proportional to the number of cases and deaths of colorectal cancer. Nanoparticles (NPs), the nanomedical drug delivery system (NDDS) is a hot topic in the field. Photodynamic therapy (PDT), immunogenic cell death (ICD), tumor microenvironment (TEM), folic acid, and pH are the cutting edge of the field. Conclusion This paper introduces the research hotspot, emphasis, and frontier of CRC and nanomedicine, and points out the direction for this field.
Collapse
Affiliation(s)
| | - Min Chen
- Proctology Department, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Nguyen HV, Campbell K, Painter GF, Young SL, Walker GF. Redox-responsive CpG-dextran conjugate enhances anti-tumour immunity following intratumoral administration. Int J Pharm 2024; 664:124621. [PMID: 39182745 DOI: 10.1016/j.ijpharm.2024.124621] [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: 03/31/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
Conjugation of a therapeutic agent to a polymer for enhanced delivery into target cells followed by its intracellular triggered release has proved to be an effective drug delivery approach. This approach is applied to the delivery of the immune-stimulatory unmethylated cytosine-phosphate-guanine (CpG) oligonucleotide for an anti-tumour immune response after intratumoral administration. On average four CpG-1668 molecules were covalently linked to a 40-kDa amino-functionalised dextran polymer via either a non-reversible (CpG-dextran) or an intracellular redox-responsive disulfide linkage (CpG-SS-dextran). Dynamic light scattering analysis showed that both conjugates had a similar particle size and surface charge of 17 nm and -10 mV, respectively. Agarose gel electrophoresis analysis showed that CpG-SS-dextran was stable in the extracellular low glutathione (GSH) concentration range (i.e. 10-20 μM) and was cleaved at the higher intracellular GSH concentration (5 mM), while CpG-dextran was stable in both GSH concentrations. Uptake and activation assays on bone-marrow-derived dendritic cells showed no significant difference between free CpG, CpG-dextran and CpG-SS-dextran. In a mouse subcutaneous colorectal tumour model the CpG-SS-dextran showed a statistically significantly greater inhibition of tumour growth (p < 0.03) and prolonged survival (p < 0.001) compared to CpG-dextran or free CpG. These results demonstrate that the redox-triggered intracellular release of CpG from a dextran polymer carrier has promise for intratumoral therapeutic vaccination against cancer.
Collapse
Affiliation(s)
- Hien V Nguyen
- Faculty of Pharmacy, Van Lang University, Ho Chi Minh City 70000, Vietnam; School of Pharmacy, University of Otago, Dunedin 9016, New Zealand.
| | - Katrin Campbell
- Department of Pathology, University of Otago, Dunedin 9016, New Zealand
| | - Gavin F Painter
- The Ferrier Research Institute, Victoria University of Wellington, Wellington 5040, New Zealand.
| | - Sarah L Young
- Faculty of Science, University of Canterbury, Christchurch 8140, New Zealand.
| | - Greg F Walker
- School of Pharmacy, University of Otago, Dunedin 9016, New Zealand.
| |
Collapse
|
5
|
Deng Z, Qing Q, Huang B. A bibliometric analysis of the application of the PI3K-AKT-mTOR signaling pathway in cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7255-7272. [PMID: 38709265 DOI: 10.1007/s00210-024-03112-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
PI3K-AKT-mTOR plays as important role in the growth, metabolism, proliferation, and migration of cancer cells, and in apoptosis, autophagy, inflammation, and angiogenesis in cancer. In this study, the aim was to comprehensively review the current research landscape regarding the PI3K-AKT-mTOR pathway in cancer, using bibliometrics to analyze research hotspots, and provide ideas for future research directions. Literature published on the topic between January 2006 and May 2023 was retrieved from the Web of Science core database, and key information and a visualization map were analyzed using CiteSpace and VOSviewer. A total of 5800 articles from 95 countries/regions were collected, including from China and the USA. The number of publications on the topic increased year on year. The major research institution was the University of Texas MD Anderson Cancer Center. Oncotarget and Clinical Cancer Research were the most prevalent journals in the field. Of 26,621 authors, R Kurzrock published the most articles, and J Engelman was cited most frequently. "A549 cell," "first line treatment," "first in human phase I," and "inhibitor" were the keywords of emerging research hotspots. Inhibitors of the PI3K-AKT-mTOR pathway and their use in clinical therapeutic strategies for cancer were the main topics in the field, and future research should also focus on PI3K-AKT-mTOR pathway inhibitors. This study is the first to comprehensively summarize trends and development s in research into the PI3K-AKT-mTOR pathway in cancer. The information that was obtained clarified recent research frontiers and directions, providing references for scholars of cancer management.
Collapse
Affiliation(s)
- Zhengzheng Deng
- School of Public Health, University of South China, Hengyang, 421001, Hunan Province, China
| | - Qiancheng Qing
- School of Public Health, University of South China, Hengyang, 421001, Hunan Province, China
| | - Bo Huang
- School of Public Health, University of South China, Hengyang, 421001, Hunan Province, China.
| |
Collapse
|
6
|
Yang L, Hu Q, Huang T. Breast Cancer Treatment Strategies Targeting the Tumor Microenvironment: How to Convert "Cold" Tumors to "Hot" Tumors. Int J Mol Sci 2024; 25:7208. [PMID: 39000314 PMCID: PMC11241188 DOI: 10.3390/ijms25137208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Breast cancer characterized as "cold tumors" exhibit low levels of immune cell infiltration, which limits the efficacy of conventional immunotherapy. Recent studies have focused on strategies using nanotechnology combined with tumor microenvironment modulation to transform "cold tumors" into "hot tumors". This approach involves the use of functionalized nanoparticles that target and modify the tumor microenvironment to promote the infiltration and activation of antitumor immune cells. By delivering immune activators or blocking immunosuppressive signals, these nanoparticles activate otherwise dormant immune responses, enhancing tumor immunogenicity and the therapeutic response. These strategies not only promise to increase the response rate of breast cancer patients to existing immunotherapies but also may pave new therapeutic avenues, providing a new direction for the immunotherapy of breast cancer.
Collapse
Affiliation(s)
- Liucui Yang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qingyi Hu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
7
|
Jiang Z, Fu Y, Shen H. Development of Intratumoral Drug Delivery Based Strategies for Antitumor Therapy. Drug Des Devel Ther 2024; 18:2189-2202. [PMID: 38882051 PMCID: PMC11179649 DOI: 10.2147/dddt.s467835] [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: 03/26/2024] [Accepted: 05/23/2024] [Indexed: 06/18/2024] Open
Abstract
Research for tumor treatment with significant therapy effects and minimal side-effects has been widely carried over the past few decades. Different drug forms have received a lot of attention. However, systemic biodistribution induces efficacy and safety issues. Intratumoral delivery of agents might overcome these problems because of its abundant tumor accumulation and retention, thereby reducing side effects. Delivering hydrogels, nanoparticles, microneedles, and microspheres drug carriers directly to tumors can realize not only targeted tumor therapy but also low side-effects. Furthermore, intratumoral administration has been integrated with treatment strategies such as chemotherapy, enhancing radiotherapy, immunotherapy, phototherapy, magnetic fluid hyperthermia, and multimodal therapy. Some of these strategies are ongoing clinical trials or applied clinically. However, many barriers hinder it from being an ideal and widely used option, such as decreased drug penetration impeded by collagen fibers of a tumor, drug squeezed out by high density and high pressure, mature intratumoral injection technique. In this review, we systematically discuss intratumoral delivery of different drug carriers and current development of intratumoral therapy strategies.
Collapse
Affiliation(s)
- Zhimei Jiang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, People’s Republic of China
| | - Yuzhi Fu
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, People’s Republic of China
| | - Hongxin Shen
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Evidence-Based Pharmacy Center, West China Second University Hospital of Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, People’s Republic of China
| |
Collapse
|
8
|
Mantooth SM, Abdou Y, Saez-Ibañez AR, Upadhaya S, Zaharoff DA. Intratumoral delivery of immunotherapy to treat breast cancer: current development in clinical and preclinical studies. Front Immunol 2024; 15:1385484. [PMID: 38803496 PMCID: PMC11128577 DOI: 10.3389/fimmu.2024.1385484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Breast cancer poses one of the largest threats to women's health. Treatment continues to improve for all the subtypes of breast cancer, but some subtypes, such as triple negative breast cancer, still present a significant treatment challenge. Additionally, metastasis and local recurrence are two prevalent problems in breast cancer treatment. A newer type of therapy, immunotherapy, may offer alternatives to traditional treatments for difficult-to-treat subtypes. Immunotherapy engages the host's immune system to eradicate disease, with the potential to induce long-lasting, durable responses. However, systemic immunotherapy is only approved in a limited number of indications, and it benefits only a minority of patients. Furthermore, immune related toxicities following systemic administration of potent immunomodulators limit dosing and, consequently, efficacy. To address these safety considerations and improve treatment efficacy, interest in local delivery at the site of the tumor has increased. Numerous intratumorally delivered immunotherapeutics have been and are being explored clinically and preclinically, including monoclonal antibodies, cellular therapies, viruses, nucleic acids, cytokines, innate immune agonists, and bacteria. This review summarizes the current and past intratumoral immunotherapy clinical landscape in breast cancer as well as current progress that has been made in preclinical studies, with a focus on delivery parameters and considerations.
Collapse
Affiliation(s)
- Siena M. Mantooth
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
| | - Yara Abdou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | | | - David A. Zaharoff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
9
|
Sandhu ZA, Raza MA, Alqurashi A, Sajid S, Ashraf S, Imtiaz K, Aman F, Alessa AH, Shamsi MB, Latif M. Advances in the Optimization of Fe Nanoparticles: Unlocking Antifungal Properties for Biomedical Applications. Pharmaceutics 2024; 16:645. [PMID: 38794307 PMCID: PMC11124843 DOI: 10.3390/pharmaceutics16050645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
In recent years, nanotechnology has achieved a remarkable status in shaping the future of biological applications, especially in combating fungal diseases. Owing to excellence in nanotechnology, iron nanoparticles (Fe NPs) have gained enormous attention in recent years. In this review, we have provided a comprehensive overview of Fe NPs covering key synthesis approaches and underlying working principles, the factors that influence their properties, essential characterization techniques, and the optimization of their antifungal potential. In addition, the diverse kinds of Fe NP delivery platforms that command highly effective release, with fewer toxic effects on patients, are of great significance in the medical field. The issues of biocompatibility, toxicity profiles, and applications of optimized Fe NPs in the field of biomedicine have also been described because these are the most significant factors determining their inclusion in clinical use. Besides this, the difficulties and regulations that exist in the transition from laboratory to experimental clinical studies (toxicity, specific standards, and safety concerns) of Fe NPs-based antifungal agents have been also summarized.
Collapse
Affiliation(s)
- Zeshan Ali Sandhu
- Department of Chemistry, Faculty of Science, Hafiz Hayat Campus, University of Gujrat, Gujrat 50700, Pakistan; (Z.A.S.); (S.A.); (K.I.)
| | - Muhammad Asam Raza
- Department of Chemistry, Faculty of Science, Hafiz Hayat Campus, University of Gujrat, Gujrat 50700, Pakistan; (Z.A.S.); (S.A.); (K.I.)
| | - Abdulmajeed Alqurashi
- Department of Biology, College of Science, Taibah University, Madinah 42353, Saudi Arabia;
| | - Samavia Sajid
- Department of Chemistry, Faculty of Science, University of Engineering and Technology, Lahore 54890, Pakistan;
| | - Sufyan Ashraf
- Department of Chemistry, Faculty of Science, Hafiz Hayat Campus, University of Gujrat, Gujrat 50700, Pakistan; (Z.A.S.); (S.A.); (K.I.)
| | - Kainat Imtiaz
- Department of Chemistry, Faculty of Science, Hafiz Hayat Campus, University of Gujrat, Gujrat 50700, Pakistan; (Z.A.S.); (S.A.); (K.I.)
| | - Farhana Aman
- Department of Chemistry, The University of Lahore, Sargodha Campus, Sargodha 40100, Pakistan;
| | - Abdulrahman H. Alessa
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Monis Bilal Shamsi
- Centre for Genetics and Inherited Diseases (CGID), Taibah University, Madinah 42353, Saudi Arabia;
- Department Basic Medical Sciences, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia
| | - Muhammad Latif
- Centre for Genetics and Inherited Diseases (CGID), Taibah University, Madinah 42353, Saudi Arabia;
- Department Basic Medical Sciences, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia
| |
Collapse
|
10
|
Lafuente-Gómez N, de Lázaro I, Dhanjani M, García-Soriano D, Sobral MC, Salas G, Mooney DJ, Somoza Á. Multifunctional magnetic nanoparticles elicit anti-tumor immunity in a mouse melanoma model. Mater Today Bio 2023; 23:100817. [PMID: 37822453 PMCID: PMC10562177 DOI: 10.1016/j.mtbio.2023.100817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/13/2023] Open
Abstract
Immunotherapy has emerged as a promising strategy to eradicate cancer cells. Particularly, the development of cancer vaccines to induce a potent and sustained antigen-specific T cell response has become a center of attention. Herein, we describe a novel immunotherapy based on magnetic nanoparticles (MNP) covalently modified with the OVA254-267 antigen and a CpG oligonucleotide via disulfide bonds. The MNP-CpG-COVA significantly enhances dendritic cell activation and CD8+ T cell antitumoral response against B16-OVA melanoma cells in vitro. Notably, the immune response induced by the covalently modified MNP is more potent and sustained over time than that triggered by the free components, highlighting the advantage of nanoformulations in immunotherapies. What is more, the nanoparticles are stable in the blood after in vivo administration and induce potent levels of systemic tumor-specific effector CD8 + T cells. Overall, our findings highlight the potential of covalently functionalized MNP to induce robust immune responses against mouse melanoma.
Collapse
Affiliation(s)
- Nuria Lafuente-Gómez
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Immunology Service, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa, Madrid, 28006, Spain
| | - Irene de Lázaro
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, New York University, New York, NY, 10010, USA
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York University, New York, NY, 10010, USA
| | - Mónica Dhanjani
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
| | - David García-Soriano
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
| | - Miguel C. Sobral
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Gorka Salas
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Unidad de Nanobiotecnología Asociada al Centro Nacional de Biotecnología (CNB-CSIC), Madrid, 28049, Spain
| | - David J. Mooney
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Unidad de Nanobiotecnología Asociada al Centro Nacional de Biotecnología (CNB-CSIC), Madrid, 28049, Spain
| |
Collapse
|
11
|
Sudduth ER, Trautmann-Rodriguez M, Gill N, Bomb K, Fromen CA. Aerosol pulmonary immune engineering. Adv Drug Deliv Rev 2023; 199:114831. [PMID: 37100206 PMCID: PMC10527166 DOI: 10.1016/j.addr.2023.114831] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/23/2023] [Accepted: 04/14/2023] [Indexed: 04/28/2023]
Abstract
Aerosolization of immunotherapies poses incredible potential for manipulating the local mucosal-specific microenvironment, engaging specialized pulmonary cellular defenders, and accessing mucosal associated lymphoid tissue to redirect systemic adaptive and memory responses. In this review, we breakdown key inhalable immunoengineering strategies for chronic, genetic, and infection-based inflammatory pulmonary disorders, encompassing the historic use of immunomodulatory agents, the transition to biological inspired or derived treatments, and novel approaches of complexing these materials into drug delivery vehicles for enhanced release outcomes. Alongside a brief description of key immune targets, fundamentals of aerosol drug delivery, and preclinical pulmonary models for immune response, we survey recent advances of inhaled immunotherapy platforms, ranging from small molecules and biologics to particulates and cell therapies, as well as prophylactic vaccines. In each section, we address the formulation design constraints for aerosol delivery as well as advantages for each platform in driving desirable immune modifications. Finally, prospects of clinical translation and outlook for inhaled immune engineering are discussed.
Collapse
Affiliation(s)
- Emma R Sudduth
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | | | - Nicole Gill
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Kartik Bomb
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Catherine A Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
12
|
Xiao M, Tang Q, Zeng S, Yang Q, Yang X, Tong X, Zhu G, Lei L, Li S. Emerging biomaterials for tumor immunotherapy. Biomater Res 2023; 27:47. [PMID: 37194085 DOI: 10.1186/s40824-023-00369-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/23/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND The immune system interacts with cancer cells in various intricate ways that can protect the individual from overproliferation of cancer cells; however, these interactions can also lead to malignancy. There has been a dramatic increase in the application of cancer immunotherapy in the last decade. However, low immunogenicity, poor specificity, weak presentation efficiency, and off-target side effects still limit its widespread application. Fortunately, advanced biomaterials effectively contribute immunotherapy and play an important role in cancer treatment, making it a research hotspot in the biomedical field. MAIN BODY This review discusses immunotherapies and the development of related biomaterials for application in the field. The review first summarizes the various types of tumor immunotherapy applicable in clinical practice as well as their underlying mechanisms. Further, it focuses on the types of biomaterials applied in immunotherapy and related research on metal nanomaterials, silicon nanoparticles, carbon nanotubes, polymer nanoparticles, and cell membrane nanocarriers. Moreover, we introduce the preparation and processing technologies of these biomaterials (liposomes, microspheres, microneedles, and hydrogels) and summarize their mechanisms when applied to tumor immunotherapy. Finally, we discuss future advancements and shortcomings related to the application of biomaterials in tumor immunotherapy. CONCLUSION Research on biomaterial-based tumor immunotherapy is booming; however, several challenges remain to be overcome to transition from experimental research to clinical application. Biomaterials have been optimized continuously and nanotechnology has achieved continuous progression, ensuring the development of more efficient biomaterials, thereby providing a platform and opportunity for breakthroughs in tumor immunotherapy.
Collapse
Affiliation(s)
- Minna Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shiying Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinying Tong
- Department of Hemodialysis, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
13
|
Liu M, Zhao Y, Shi Z, Zink JI, Yu Q. Virus-like Magnetic Mesoporous Silica Particles as a Universal Vaccination Platform against Pathogenic Infections. ACS NANO 2023; 17:6899-6911. [PMID: 36961475 DOI: 10.1021/acsnano.3c00644] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Vaccination is the most important way of population protection from life-threatening pathogenic infections. However, its efficiency is frequently compromised by a failure of strong antigen presentation and immune activation. Herein, we developed virus-like magnetic mesoporous silica nanoparticles as a universal vaccination platform (termed MagParV) for preventing pathogenic infections. This platform was constructed by integrating synthetic biology-based endoplasmic reticulum-targeting vesicles with magnetic mesoporous silica particles. This platform exhibited high antigen-loading capacity, strongly targeting the endoplasmic reticulum and promoting antigen presentation in dendritic cells. After prime-boost vaccination, the antigen-loading MagParV with AMF drastically elicited specific antibody production against corresponding antigens of fungal, bacterial, and viral pathogens. A systemic infection model further revealed that the platform effectively protected the mice from severe fungal systemic infections. This study realized synthetic biology-facilitated green manufacturing of vaccines, which is promising for magnetism-activated vaccination against different kinds of pathogenic infections.
Collapse
Affiliation(s)
- Mingyang Liu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, People's Republic of China
| | - Yan Zhao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Zhishang Shi
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Jeffrey I Zink
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- California Nano Systems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| |
Collapse
|
14
|
Nanomaterials: Breaking through the bottleneck of tumor immunotherapy. Int J Biol Macromol 2023; 230:123159. [PMID: 36610572 DOI: 10.1016/j.ijbiomac.2023.123159] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/23/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
Immunotherapy exerts its excellent anti-tumor effects by stimulating and enhancing the immune response of the body, and has become another important class of anti-tumor therapy besides chemotherapy, targeted therapy and radiotherapy. Various types of immunotherapeutic drugs have gained their clinical values, but the in vivo delivery of drugs still faces many challenges, such as poor tumor permeability and low tumor cell uptake rate. In recent years, owing to highly targeting properties, better biocompatibility, and easy functionalization, nanomaterials have been widely applicated in tumor treatment, especially in tumor immunotherapy. Furthermore, nanomaterials have large drug loading capacity, strong tumor targeting and easy modification, which can effectively overcome the drawbacks of traditional immunotherapy. This paper reviews the progress of nanomaterial-based tumor immunotherapy in recent years and provides a theoretical basis for exploring new nanomaterial-based tumor immunotherapy strategies.
Collapse
|
15
|
Yan B, Wang S, Liu C, Wen N, Li H, Zhang Y, Wang H, Xi Z, Lv Y, Fan H, Liu X. Engineering magnetic nano-manipulators for boosting cancer immunotherapy. J Nanobiotechnology 2022; 20:547. [PMID: 36587223 PMCID: PMC9805281 DOI: 10.1186/s12951-022-01760-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023] Open
Abstract
Cancer immunotherapy has shown promising therapeutic results in the clinic, albeit only in a limited number of cancer types, and its efficacy remains less than satisfactory. Nanoparticle-based approaches have been shown to increase the response to immunotherapies to address this limitation. In particular, magnetic nanoparticles (MNPs) as a powerful manipulator are an appealing option for comprehensively regulating the immune system in vivo due to their unique magnetically responsive properties and high biocompatibility. This review focuses on assessing the potential applications of MNPs in enhancing tumor accumulation of immunotherapeutic agents and immunogenicity, improving immune cell infiltration, and creating an immunotherapy-sensitive environment. We summarize recent progress in the application of MNP-based manipulators to augment the efficacy of immunotherapy, by MNPs and their multiple magnetically responsive effects under different types of external magnetic field. Furthermore, we highlight the mechanisms underlying the promotion of antitumor immunity, including magnetically actuated delivery and controlled release of immunotherapeutic agents, tracking and visualization of immune response in real time, and magnetic regulation of innate/adaptive immune cells. Finally, we consider perspectives and challenges in MNP-based immunotherapy.
Collapse
Affiliation(s)
- Bin Yan
- grid.412262.10000 0004 1761 5538Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, 710069 Shaanxi China
| | - Siyao Wang
- grid.412262.10000 0004 1761 5538Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, 710069 Shaanxi China
| | - Chen Liu
- grid.412262.10000 0004 1761 5538Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, 710069 Shaanxi China
| | - Nana Wen
- grid.412262.10000 0004 1761 5538Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, 710069 Shaanxi China
| | - Hugang Li
- grid.412262.10000 0004 1761 5538Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, 710069 Shaanxi China
| | - Yihan Zhang
- grid.412262.10000 0004 1761 5538College of Chemistry & Materials Science, Northwest University, Xi’an, 710127 Shaanxi China
| | - Hao Wang
- grid.412262.10000 0004 1761 5538Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, 710069 Shaanxi China
| | - Ziyi Xi
- grid.412262.10000 0004 1761 5538Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, 710069 Shaanxi China
| | - Yi Lv
- grid.452438.c0000 0004 1760 8119Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China ,grid.452438.c0000 0004 1760 8119National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi China
| | - Haiming Fan
- grid.412262.10000 0004 1761 5538Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, 710069 Shaanxi China ,grid.412262.10000 0004 1761 5538College of Chemistry & Materials Science, Northwest University, Xi’an, 710127 Shaanxi China
| | - Xiaoli Liu
- grid.412262.10000 0004 1761 5538Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, 710069 Shaanxi China ,grid.452438.c0000 0004 1760 8119Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China ,grid.452438.c0000 0004 1760 8119National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi China
| |
Collapse
|
16
|
Pei Z, Chen S, Ding L, Liu J, Cui X, Li F, Qiu F. Current perspectives and trend of nanomedicine in cancer: A review and bibliometric analysis. J Control Release 2022; 352:211-241. [PMID: 36270513 DOI: 10.1016/j.jconrel.2022.10.023] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022]
Abstract
The limitations of traditional cancer treatments are driving the creation and development of new nanomedicines. At present, with the rapid increase of research on nanomedicine in the field of cancer, there is a lack of intuitive analysis of the development trend, main authors and research hotspots of nanomedicine in the field of cancer, as well as detailed elaboration of possible research hotspots. In this review, data collected from the Web of Science Core Collection database between January 1st, 2000, and December 31st, 2021, were subjected to a bibliometric analysis. The co-authorship, co-citation, and co-occurrence of countries, institutions, authors, literature, and keywords in this subject were examined using VOSviewer, Citespace, and a well-known online bibliometrics platform. We collected 19,654 published papers, China produced the most publications (36.654%, 7204), followed by the United States (29.594%, 5777), and India (7.780%, 1529). An interesting fact is that, despite China having more publications than the United States, the United States still dominates this field, having the highest H-index and the most citations. Acs Nano, Nano Letters, and Biomaterials are the top three academic publications that publish articles on nanomedicine for cancer out of a total of 7580 academic journals. The most significant increases were shown for the keywords "cancer nanomedicine", "tumor microenvironment", "nanoparticles", "prodrug", "targeted nanomedicine", "combination", and "cancer immunotherapy" indicating the promising area of research. Meanwhile, the development prospects and challenges of nanomedicine in cancer are also discussed and provided some solutions to the major obstacles.
Collapse
Affiliation(s)
- Zerong Pei
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuting Chen
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Liqin Ding
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingbo Liu
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Xinyi Cui
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Fengyun Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
17
|
Zhang J, Zhang T, Gao J. Biocompatible Iron Oxide Nanoparticles for Targeted Cancer Gene Therapy: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12193323. [PMID: 36234452 PMCID: PMC9565336 DOI: 10.3390/nano12193323] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 05/14/2023]
Abstract
In recent years, gene therapy has made remarkable achievements in tumor treatment. In a successfully cancer gene therapy, a smart gene delivery system is necessary for both protecting the therapeutic genes in circulation and enabling high gene expression in tumor sites. Magnetic iron oxide nanoparticles (IONPs) have demonstrated their bright promise for highly efficient gene delivery target to tumor tissues, partly due to their good biocompatibility, magnetic responsiveness, and extensive functional surface modification. In this review, the latest progress in targeting cancer gene therapy is introduced, and the unique properties of IONPs contributing to the efficient delivery of therapeutic genes are summarized with detailed examples. Furthermore, the diagnosis potentials and synergistic tumor treatment capacity of IONPs are highlighted. In addition, aiming at potential risks during the gene delivery process, several strategies to improve the efficiency or reduce the potential risks of using IONPs for cancer gene therapy are introduced and addressed. The strategies and applications summarized in this review provide a general understanding for the potential applications of IONPs in cancer gene therapy.
Collapse
Affiliation(s)
- Jinsong Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Correspondence: (T.Z.); (J.G.)
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Correspondence: (T.Z.); (J.G.)
| |
Collapse
|
18
|
Chang Q, Chang L, Li M, Fan L, Bao S, Wang X, Liu L. Nanobiotherapeutic strategies to target immune microenvironment of triple-negative breast cancer. Am J Cancer Res 2022; 12:4083-4102. [PMID: 36225648 PMCID: PMC9548023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/23/2022] [Indexed: 06/16/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the subtype with the least favourable outcomes in breast cancer. Besides chemotherapy, there is a chronic lack of other effective treatments. Advances in omic technologies have liberated us from the ambiguity of TNBC heterogeneity in terms of cancer cell and immune microenvironment in recent years. This new understanding of TNBC pathology has already led to the exploitation of novel nanoparticulate systems, including tumor vaccines, oncolytic viruses, and antibody derivatives. The revolutionary ideas in the therapeutic landscape provide new opportunities for TNBC patients. Translating these experimental medicines into clinical benefit is both appreciated and challenging. In this review, we describe the prospective nanobiotherapy of TNBC that has been developed to overcome clinical obstacles, and provide our vision for this booming field at the overlap of cancer biotherapy and nanomaterial design.
Collapse
Affiliation(s)
- Qing Chang
- Department of Radiotherapy, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin, China
- Jilin Provincial Key Laboratory of Early Screening and Health Management for Cancer, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin, China
- Biotechnology and Medical Materials Engineering Research Center of Jilin Province, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin, China
| | - Liang Chang
- Xi’an Technological UniversityXi’an, Shanxi, China
| | - Mo Li
- The Second Hospital of Jilin UniversityChangchun, Jilin, China
| | - Liwen Fan
- Department of Radiotherapy, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin, China
| | - Shunchao Bao
- Department of Radiotherapy, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin, China
| | - Xinyu Wang
- The Second Hospital of Jilin UniversityChangchun, Jilin, China
| | - Linlin Liu
- Department of Radiotherapy, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin, China
- Jilin Provincial Key Laboratory of Early Screening and Health Management for Cancer, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin, China
- Biotechnology and Medical Materials Engineering Research Center of Jilin Province, China-Japan Union Hospital of Jilin UniversityChangchun, Jilin, China
| |
Collapse
|
19
|
Haegebaert RM, Kempers M, Ceelen W, Lentacker I, Remaut K. Nanoparticle mediated targeting of toll-like receptors to treat colorectal cancer. Eur J Pharm Biopharm 2022; 172:16-30. [PMID: 35074555 DOI: 10.1016/j.ejpb.2022.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
|
20
|
Darroudi M, Gholami M, Rezayi M, Khazaei M. An overview and bibliometric analysis on the colorectal cancer therapy by magnetic functionalized nanoparticles for the responsive and targeted drug delivery. J Nanobiotechnology 2021; 19:399. [PMID: 34844632 PMCID: PMC8630862 DOI: 10.1186/s12951-021-01150-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/19/2021] [Indexed: 12/27/2022] Open
Abstract
With the growing demands for personalized medicine and medical devices, nanomedicine is a modern scientific field, and research continues to apply nanomaterials for therapeutic and damaged tissue diagnosis. In this regard, substantial progress has been made in synthesizing magnetic nanoparticles with desired sizes, chemical composition, morphologies, and surface chemistry. Among these materials, nanomagnetic iron oxides have demonstrated promise as unique drug delivery carriers due to cancer treatment. This carrier could lead to responsive properties to a specific trigger, including heat, pH, alternative magnetic field, or even enzymes, through functionalization and coating of magnetic nanoparticles, along with biocompatibility, good chemical stability, easy functionalization, simple processing, and ability to localize to the tumor site with the assistance of external magnetic field. Current studies have focused on magnetic nanoparticles' utilities in cancer therapy, especially for colorectal cancer. Additionally, a bibliometric investigation was performed on the public trends in the field of the magnetic nanoparticle to drug delivery and anticancer, which represented progressing applications of these carriers in the multidisciplinary zones with a general view on future research and identified potential opportunities and challenges. Furthermore, we outline the current challenges and forthcoming research perspective for high performance and fostering advanced MNPs in colorectal cancer treatment.
Collapse
Affiliation(s)
- Mahdieh Darroudi
- Department of Medical Biotechnology and Nanotechnology, School of Science, Mashhad University of Medical Science, Mashhad, Iran.,Department of Physiology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Mehrdad Gholami
- Department of Chemistry, Marvdasht Branch, Islamic Azad University, P.O. Box 465, Marvdasht, Iran
| | - Majid Rezayi
- Department of Medical Biotechnology and Nanotechnology, School of Science, Mashhad University of Medical Science, Mashhad, Iran. .,Medical Toxicology Research Center, Mashhad University of Medical Science, Mashhad, Iran. .,Metabolic Syndrome Research Center, Mashhad University of Medical Science, Mashhad, Iran.
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran. .,Metabolic Syndrome Research Center, Mashhad University of Medical Science, Mashhad, Iran.
| |
Collapse
|
21
|
Li J, Men K, Gao Y, Wu J, Lei S, Yang Y, Pan H. Single Micelle Vectors based on Lipid/Block Copolymer Compositions as mRNA Formulations for Efficient Cancer Immunogene Therapy. Mol Pharm 2021; 18:4029-4045. [PMID: 34559545 DOI: 10.1021/acs.molpharmaceut.1c00461] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Immunogene therapy provides a new strategy for the treatment of colorectal cancer. Compared to plasmid DNA, mRNA possesses several advantages as a therapeutic nucleic acid material and shows high potential in cancer therapy. Although efforts have been made to conquer the limited efficiency of mRNA delivery, most of the current mRNA vectors possess complex structures or compositions, which introduces additional toxicity and hinders their further clinical application. Hence, it is highly necessary to develop potent mRNA delivery systems with simple structures. Here, we report efficient mRNA delivery using the biodegradable micelle delivery system of DMP (DOTAP-mPEG-PCL). Biodegradable DMP micelles were simply prepared by the self-assembly of cationic lipid DOTAP and the diblock polymer monomethoxy poly(ethylene glycol)-poly(ε-caprolactone). With an average size of only 30 nm, we proved that these single-structured cationic micelles are highly potent in condensing and protecting mRNA molecules, with a delivery efficiency of 60.59% on C26 mouse colon cancer cells. The micelles triggered specific internalization pathways and were fully degraded in vivo. After binding with IL-22BP (interleukin-22 binding protein)-encoding mRNA, a strongly elevated IL-22BP mRNA level was detected in C26 cells. After intraperitoneal and intratumoral injection of the DMP/mIL-22BP complex, strong inhibition effects on C26 colon cancer models were observed, with high therapeutic efficiency and safety when systemically administrated. These data suggest that the DMP micelle is an advanced single-structured mRNA delivery system with high safety.
Collapse
Affiliation(s)
- Jingmei Li
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yan Gao
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jieping Wu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Sibei Lei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Haixia Pan
- Oncology Center, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China
| |
Collapse
|
22
|
Shukla A, Cano-Mejia J, Andricovich J, Burga RA, Sweeney EE, Fernandes R. An Engineered Prussian Blue Nanoparticles-based Nanoimmunotherapy Elicits Robust and Persistent Immunological Memory in a TH-MYCN Neuroblastoma Model. ADVANCED NANOBIOMED RESEARCH 2021; 1. [PMID: 34435194 DOI: 10.1002/anbr.202100021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A combination therapy using Prussian blue nanoparticles (PBNP) as photothermal therapy (PTT) agents coated with CpG oligodeoxynucleotides, an immunologic adjuvant, as a nanoimmunotherapy (CpG-PBNP-PTT) for neuroblastoma (NB) is described. NB driven by MYCN amplification confers high risk and correlates with a dismal prognosis, accounting for the majority of NB-related mortality. The efficacy of the CpG-PBNP-PTT nanoimmunotherapy in a clinically relevant, TH-MYCN murine NB model (9464D) overexpressing MYCN is tested. When administered to 9464D NB cells in vitro, CpG-PBNP-PTT triggers thermal dose-dependent immunogenic cell death and tumor cell priming for immune recognition in vitro, measured by the expression of specific costimulatory and antigen-presenting molecules. In vivo, intratumorally administered CpG-PBNP-PTT generates complete tumor regression and significantly higher long-term survival compared to controls. Furthermore, CpG-PBNP-PTT-treated mice reject tumor rechallenge. Ex vivo studies confirm these therapeutic responses result from the generation of robust T cell-mediated immunological memory. Consequently, in a synchronous 9464D tumor model, CpG-PBNP-PTT induces complete tumor regression on the treated flank and significantly slows tumor progression on the untreated flank, improving animal survival. These findings demonstrate that localized administration of the CpG-PBNP-PTT nanoimmunotherapy drives potent systemic T cell responses in solid tumors such as NB and therefore has therapeutic implications for NB.
Collapse
Affiliation(s)
- Anshi Shukla
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| | - Juliana Cano-Mejia
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| | - Jaclyn Andricovich
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA.,The Institute for Biomedical Sciences, The George Washington University,2300 Eye Street NW, Ross Hall Room 561, Washington, DC 20037, USA
| | - Rachel A Burga
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA.,The Institute for Biomedical Sciences, The George Washington University,2300 Eye Street NW, Ross Hall Room 561, Washington, DC 20037, USA
| | - Elizabeth E Sweeney
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| | - Rohan Fernandes
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| |
Collapse
|
23
|
Day NB, Wixson WC, Shields CW. Magnetic systems for cancer immunotherapy. Acta Pharm Sin B 2021; 11:2172-2196. [PMID: 34522583 PMCID: PMC8424374 DOI: 10.1016/j.apsb.2021.03.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/05/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy is a rapidly developing area of cancer treatment due to its higher specificity and potential for greater efficacy than traditional therapies. Immune cell modulation through the administration of drugs, proteins, and cells can enhance antitumoral responses through pathways that may be otherwise inhibited in the presence of immunosuppressive tumors. Magnetic systems offer several advantages for improving the performance of immunotherapies, including increased spatiotemporal control over transport, release, and dosing of immunomodulatory drugs within the body, resulting in reduced off-target effects and improved efficacy. Compared to alternative methods for stimulating drug release such as light and pH, magnetic systems enable several distinct methods for programming immune responses. First, we discuss how magnetic hyperthermia can stimulate immune cells and trigger thermoresponsive drug release. Second, we summarize how magnetically targeted delivery of drug carriers can increase the accumulation of drugs in target sites. Third, we review how biomaterials can undergo magnetically driven structural changes to enable remote release of encapsulated drugs. Fourth, we describe the use of magnetic particles for targeted interactions with cellular receptors for promoting antitumor activity. Finally, we discuss translational considerations of these systems, such as toxicity, clinical compatibility, and future opportunities for improving cancer treatment.
Collapse
Key Words
- BW, body weight
- Biomaterials
- CpG, cytosine-phosphate-guanine
- DAMP, damage associated molecular pattern
- Drug delivery
- EPR, enhanced permeability and retention
- FFR, field free region
- HS-TEX, heat-stressed tumor cell exosomes
- HSP, heat shock protein
- ICD, immunogenic cell death
- IVIS, in vivo imaging system
- Immunotherapy
- MICA, MHC class I-related chain A
- MPI, magnetic particle imaging
- Magnetic hyperthermia
- Magnetic nanoparticles
- Microrobotics
- ODNs, oligodeoxynucleotides
- PARP, poly(adenosine diphosphate-ribose) polymerase
- PDMS, polydimethylsiloxane
- PEG, polyethylene glycol
- PLGA, poly(lactic-co-glycolic acid)
- PNIPAM, poly(N-isopropylacrylamide)
- PVA, poly(vinyl alcohol)
- SDF, stromal cell derived-factor
- SID, small implantable device
- SLP, specific loss power
Collapse
Affiliation(s)
- Nicole B Day
- Department of Chemical & Biological Engineering, University of Colorado, Boulder, CO 80303, USA
| | - William C Wixson
- Department of Chemical & Biological Engineering, University of Colorado, Boulder, CO 80303, USA
| | - C Wyatt Shields
- Department of Chemical & Biological Engineering, University of Colorado, Boulder, CO 80303, USA
| |
Collapse
|
24
|
Gorbet MJ, Singh A, Mao C, Fiering S, Ranjan A. Using nanoparticles for in situ vaccination against cancer: mechanisms and immunotherapy benefits. Int J Hyperthermia 2021; 37:18-33. [PMID: 33426995 DOI: 10.1080/02656736.2020.1802519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy to treat cancer is now an established clinical approach. Immunotherapy can be applied systemically, as done with checkpoint blockade antibodies, but it can also be injected directly into identified tumors, in a strategy of in situ vaccination (ISV). ISV is designed to stimulate a strong local antitumor immune response involving both innate and adaptive immune cells, and through this generate a systemic antitumor immune response against metastatic tumors. A variety of ISVs have been utilized to generate an immunostimulatory tumor microenvironment (TME). These include attenuated microorganisms, recombinant proteins, small molecules, physical disruptors of TME (alternating magnetic and focused ultrasound heating, photothermal therapy, and radiotherapy), and more recently nanoparticles (NPs). NPs are attractive and unique since they can load multiple drugs or other reagents to influence immune and cancer cell functions in the TME, affording a unique opportunity to stimulate antitumor immunity. Here, we describe the NP-ISV therapeutic mechanisms, review chemically synthesized NPs (i.e., liposomes, polymeric, chitosan-based, inorganic NPs, etc.), biologically derived NPs (virus and bacteria-based NPs), and energy-activated NP-ISVs in the context of their use as local ISV. Data suggests that NP-ISVs can enhance outcomes of immunotherapeutic regimens including those utilizing tumor hyperthermia and checkpoint blockade therapies.
Collapse
Affiliation(s)
| | - Akansha Singh
- College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Chenkai Mao
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center at Dartmouth and Dartmouth Hitchcock, Lebanon, NH, USA
| | - Ashish Ranjan
- College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| |
Collapse
|
25
|
Nano-delivery systems focused on tumor microenvironment regulation and biomimetic strategies for treatment of breast cancer metastasis. J Control Release 2021; 333:374-390. [PMID: 33798666 DOI: 10.1016/j.jconrel.2021.03.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 12/14/2022]
Abstract
Breast cancer metastasis and recurrence accounts for vast majority of breast cancer-induced mortality. Tumor microenvironment (TME) plays an important role at each step of metastasis, evasion of immunosurveillance, and therapeutic resistance. Consequently, TME-targeting alternatives to traditional therapies focused on breast cancer cells are gaining increasing attention. These new therapies involve the use of tumor cells, and key TME components or secreted bioactive molecules as therapeutic targets, alone or in combination. Recently, TME-related nanoparticles have been developed to deliver various agents, such as bioactive ingredients extracted from natural sources or chemotherapeutic agents, genes, proteins, small interfering RNAs, and vaccines; they have shown great therapeutic potential against breast cancer metastasis. Among various types of nanoparticles, biomimetic nanovesicles are a promising means of addressing the limitations of conventional nanocarriers. This review highlights various nanoparticles related to or mediated by TME according to the key TME components responsible for metastasis. Furthermore, TME-related biomimetic nanoparticles against breast cancer metastasis have garnered attention owing to their promising efficiency, especially in payload delivery and therapeutic action. Here, we summarize recent representative studies on nanoparticles related to cancer-associated fibroblasts, extracellular matrix, endothelial cells, angiogenesis, and immune cells, as well as advanced biomimetic nanoparticles. Future challenges and opportunities in the field are also discussed.
Collapse
|
26
|
Abstract
Iron-based nanomaterials have appeared in various cancer treatments owing to their promising functions and safety. Various sophisticated iron-based nanomaterials have been designed to exhibit great therapeutic effects through different strategies. Given the rapid progression, there is a great need to integrate the recent advances to learn about the latest innovation in this field. In this review, we classified the strategies of iron-based nanomaterials for cancer treatment into the following categories: immunotherapy, ferroptosis, magnetic hyperthermia and magneto-mechanical destruction. On the one hand, we discussed the underlining mechanism of iron-based nanomaterials in these therapies and applications; on the other hand, we analyzed the feasible combination of these applications and other therapies. Finally, the current challenges and expectation of iron-based nanomaterials in this field were highlighted.
Collapse
Affiliation(s)
- Xiaqing Wu
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, People's Republic of China. University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
| | | |
Collapse
|
27
|
Khan A, Dias F, Neekhra S, Singh B, Srivastava R. Designing and Immunomodulating Multiresponsive Nanomaterial for Cancer Theranostics. Front Chem 2021; 8:631351. [PMID: 33585406 PMCID: PMC7878384 DOI: 10.3389/fchem.2020.631351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/22/2020] [Indexed: 01/14/2023] Open
Abstract
Cancer has been widely investigated yet limited in its manifestation. Cancer treatment holds innovative and futuristic strategies considering high disease heterogeneity. Chemotherapy, radiotherapy and surgery are the most explored pillars; however optimal therapeutic window and patient compliance recruit constraints. Recently evolved immunotherapy demonstrates a vital role of the host immune system to prevent metastasis recurrence, still undesirable clinical response and autoimmune adverse effects remain unresolved. Overcoming these challenges, tunable biomaterials could effectively control the co-delivery of anticancer drugs and immunomodulators. Current status demands a potentially new approach for minimally invasive, synergistic, and combinatorial nano-biomaterial assisted targeted immune-based treatment including therapeutics, diagnosis and imaging. This review discusses the latest findings of engineering biomaterial with immunomodulating properties and implementing novel developments in designing versatile nanosystems for cancer theranostics. We explore the functionalization of nanoparticle for delivering antitumor therapeutic and diagnostic agents promoting immune response. Through understanding the efficacy of delivery system, we have enlightened the applicability of nanomaterials as immunomodulatory nanomedicine further advancing to preclinical and clinical trials. Future and present ongoing improvements in engineering biomaterial could result in generating better insight to deal with cancer through easily accessible immunological interventions.
Collapse
Affiliation(s)
- Amreen Khan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Centre for Research in Nanotechnology and Science, Indian Institute of Technology Bombay, Mumbai, India
| | - Faith Dias
- Department of Chemical Engineering, Thadomal Shahani Engineering College, Mumbai, India
| | - Suditi Neekhra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Barkha Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Centre for Research in Nanotechnology and Science, Indian Institute of Technology Bombay, Mumbai, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
28
|
Ahmad MZ, Ahmad J, Haque A, Alasmary MY, Abdel-Wahab BA, Akhter S. Emerging advances in synthetic cancer nano-vaccines: opportunities and challenges. Expert Rev Vaccines 2020; 19:1053-1071. [PMID: 33315512 DOI: 10.1080/14760584.2020.1858058] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Anzarul Haque
- Department of Pharmacognosy, Prince Sattam Bin Abdulaziz University College of Pharmacy, Alkharj Al-Kharj, Kingdom of Saudi Arabia
| | - Mohammed Yahia Alasmary
- Department of Internal Medicine, College of Medicine, Najran University Hospital, Najran, Kingdom of Saudi Arabia
| | - Basel A. Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
- Department of Pharmacology, College of Medicine Assiut University, Assiut, Egypt
| | - Sohail Akhter
- Center for Molecular Biophysics (CBM), CNRS UPR4301; LE STUDIUM Loire Valley Institute for Advanced Studies, Orleans, France
| |
Collapse
|
29
|
Recent advances in chemical surface modification of metal oxide nanoparticles with silane coupling agents: A review. Adv Colloid Interface Sci 2020; 286:102298. [PMID: 33171357 DOI: 10.1016/j.cis.2020.102298] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 01/22/2023]
Abstract
Nowadays, metal oxide nanoparticles (NPs) have been applied in various fields of nanotechnology including catalysis of chemical reactions, drug delivery, water treatment, textile industries, polymer composites, adhesives, and coatings. The greatest challenge in relation to metal oxide NPs is high tendency to aggregation. Chemical surface modification of metal oxide NPs has gained widely interest to control of dispersion and aggregation of NPs. Silane modifiers are one of the most important bifunctional modifiers that are frequently used for surface treatment of metal oxide NPs. In this review paper, we first focus on the synthesis, surface thermodynamic properties, surface modification techniques, and kinetic of silanization reaction of metal oxide NPs. Then, the recent development in using silane modifiers for treatment of metal oxide NPs in various applications were investigated. It was found that the unmodified NPs have high surface energy and are thermodynamically unstable. The thermodynamic properties of NPs including Gms, Hms, and Smsincrease with decreasing the particle size. Aggregation phenomena is the simple way to reduce the excess surface energy of NPs that leads to an increase in particle size. Therefore, the chemical surface modification of NPs using silane modifiers can be used as an effective method for the prevention of NPs agglomeration and improvement of NPs stability.
Collapse
|
30
|
do Nascimento T, Todeschini AR, Santos-Oliveira R, de Souza de Bustamante Monteiro MS, de Souza VT, Ricci-Júnior E. Trends in Nanomedicines for Cancer Treatment. Curr Pharm Des 2020; 26:3579-3600. [DOI: 10.2174/1381612826666200318145349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
Background:
Cancer is characterized by abnormal cell growth and considered one of the leading
causes of death around the world. Pharmaceutical Nanotechnology has been extensively studied for the optimization
of cancer treatment.
Objective:
Comprehend the panorama of Pharmaceutical Nanotechnology in cancer treatment, through a survey
about nanomedicines applied in clinical studies, approved for use and patented.
Methods:
Acknowledged products under clinical study and nanomedicines commercialized found in scientific
articles through research on the following databases: Pubmed, Science Direct, Scielo and Lilacs. Derwent tool
was used for patent research.
Results:
Nanomedicines based on nanoparticles, polymer micelles, liposomes, dendrimers and nanoemulsions
were studied, along with cancer therapies such as Photodynamic Therapy, Infrared Phototherapy Hyperthermia,
Magnetic Hyperthermia, Radiotherapy, Gene Therapy and Nanoimmunotherapy. Great advancement has been
observed over nanotechnology applied to cancer treatment, mainly for nanoparticles and liposomes.
Conclusion:
The combination of drugs in nanosystems helps to increase efficacy and decrease toxicity. Based on
the results encountered, nanoparticles and liposomes were the most commonly used nanocarriers for drug encapsulation.
In addition, although few nanomedicines are commercially available, this specific research field is continuously
growing.
Collapse
Affiliation(s)
- Tatielle do Nascimento
- Laboratorio de Desenvolvimento Galenico, Farmacia Universitária, Centro de Ciencias da Saude, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriane R. Todeschini
- Laboratorio de Glicobiologia Estrutural e Funcional, Instituto de Biofisica Carlos Chagas Filho, Centro de Ciencias da Saude, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ralph Santos-Oliveira
- Instituto de Engenharia Nuclear, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Vilênia T. de Souza
- Laboratorio de Tecnologia Industrial Farmaceutica, Centro de Ciencias da Saude, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduardo Ricci-Júnior
- Laboratorio de Desenvolvimento Galenico, Farmacia Universitária, Centro de Ciencias da Saude, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
31
|
Abdou P, Wang Z, Chen Q, Chan A, Zhou DR, Gunadhi V, Gu Z. Advances in engineering local drug delivery systems for cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1632. [PMID: 32255276 PMCID: PMC7725287 DOI: 10.1002/wnan.1632] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy aims to leverage the immune system to suppress the growth of tumors and to inhibit metastasis. The recent promising clinical outcomes associated with cancer immunotherapy have prompted research and development efforts towards enhancing the efficacy of immune checkpoint blockade, cancer vaccines, cytokine therapy, and adoptive T cell therapy. Advancements in biomaterials, nanomedicine, and micro-/nano-technology have facilitated the development of enhanced local delivery systems for cancer immunotherapy, which can enhance treatment efficacy while minimizing toxicity. Furthermore, locally administered cancer therapies that combine immunotherapy with chemotherapy, radiotherapy, or phototherapy have the potential to achieve synergistic antitumor effects. Herein, the latest studies on local delivery systems for cancer immunotherapy are surveyed, with an emphasis on the therapeutic benefits associated with the design of biomaterials and nanomedicines. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Peter Abdou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Road, Suzhou, 215123, Jiangsu, PR China
| | - Amanda Chan
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Daojia R. Zhou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Vivienne Gunadhi
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
32
|
Van Herck S, De Geest BG. Nanomedicine-mediated alteration of the pharmacokinetic profile of small molecule cancer immunotherapeutics. Acta Pharmacol Sin 2020; 41:881-894. [PMID: 32451411 PMCID: PMC7471422 DOI: 10.1038/s41401-020-0425-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022] Open
Abstract
The advent of immunotherapy is a game changer in cancer therapy with monoclonal antibody- and T cell-based therapeutics being the current flagships. Small molecule immunotherapeutics might offer advantages over the biological drugs in terms of complexity, tissue penetration, manufacturing cost, stability, and shelf life. However, small molecule drugs are prone to rapid systemic distribution, which might induce severe off-target side effects. Nanotechnology could aid in the formulation of the drug molecules to improve their delivery to specific immune cell subsets. In this review we summarize the current efforts in changing the pharmacokinetic profile of small molecule immunotherapeutics with a strong focus on Toll-like receptor agonists. In addition, we give our vision on limitations and future pathways in the route of nanomedicine to the clinical practice.
Collapse
Affiliation(s)
- Simon Van Herck
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium.
| |
Collapse
|
33
|
Huang Y, Zeng J. Recent development and applications of nanomaterials for cancer
immunotherapy. NANOTECHNOLOGY REVIEWS 2020; 9:367-384. [DOI: 10.1515/ntrev-2020-0027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Immunotherapy, which utilizes the patient’s own immune system to fight against
cancer, further results in durable antitumor responses and reduces metastasis and
recurrence, has become one of the most effective and important cancer therapies along
with surgery, radiotherapy, and chemotherapy. Nanomaterials with the advantages of
large specific surface, delivery function, and controllable surface chemistry are
used to deliver antigens or adjuvants, or both, help to boost immune responses with
the imaging function or just act as adjuvants themselves and modulate tumor
microenvironment (TME). In this review, recent development and applications of
nanomaterials for cancer immunotherapy including delivery systems based on
nanomaterials, uniting imaging, self-adjuvants, targeting functions, artificial
antigen presenting cells, and TME modulation are focused and discussed.
Collapse
Affiliation(s)
- Yao Huang
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005 , China
| | - Jinhua Zeng
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005 , China
| |
Collapse
|
34
|
Tornesello AL, Tagliamonte M, Tornesello ML, Buonaguro FM, Buonaguro L. Nanoparticles to Improve the Efficacy of Peptide-Based Cancer Vaccines. Cancers (Basel) 2020; 12:E1049. [PMID: 32340356 PMCID: PMC7226445 DOI: 10.3390/cancers12041049] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/24/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
Nanoparticles represent a potent antigen presentation and delivery system to elicit an optimal immune response by effector cells targeting tumor-associated antigens expressed by cancer cells. Many types of nanoparticles have been developed, such as polymeric complexes, liposomes, micelles and protein-based structures such as virus like particles. All of them show promising results for immunotherapy approaches. In particular, the immunogenicity of peptide-based cancer vaccines can be significantly potentiated by nanoparticles. Indeed, nanoparticles are able to enhance the targeting of antigen-presenting cells (APCs) and trigger cytokine production for optimal T cell response. The present review summarizes the categories of nanoparticles and peptide cancer vaccines which are currently under pre-clinical evaluation.
Collapse
Affiliation(s)
- Anna Lucia Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (M.L.T.); (F.M.B.)
| | - Maria Tagliamonte
- Innovative Immunological Models, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy;
| | - Maria Lina Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (M.L.T.); (F.M.B.)
| | - Franco M. Buonaguro
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (M.L.T.); (F.M.B.)
| | - Luigi Buonaguro
- Innovative Immunological Models, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy;
| |
Collapse
|
35
|
Gao J, Xia B, Li S, Huang L, Ma T, Shi X, Luo K, Yang Y, Zhao L, Zhang H, Luo B, Huang J. Magnetic Field Promotes Migration of Schwann Cells with Chondroitinase ABC (ChABC)-Loaded Superparamagnetic Nanoparticles Across Astrocyte Boundary in vitro. Int J Nanomedicine 2020; 15:315-332. [PMID: 32021182 PMCID: PMC6980842 DOI: 10.2147/ijn.s227328] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The clinical outcome of spinal cord injury is usually poor due to the lack of axonal regeneration and glia scar formation. As one of the most classical supporting cells in neural regeneration, Schwann cells (SCs) provide bioactive substrates for axonal migration and release molecules that regulate axonal growth. However, the effect of SC transplantation is limited by their poor migration capacity in the astrocyte-rich central nervous system. METHODS In this study, we first magnetofected SCs with chondroitinase ABC-polyethylenimine functionalized superparamagnetic iron oxide nanoparticles (ChABC/PEI-SPIONs) to induce overexpression of ChABC for the removal of chondroitin sulfate proteoglycans. These are inhibitory factors and forming a dense scar that acts as a barrier to the regenerating axons. In vitro, we observed the migration of SCs in the region of astrocytes after the application of a stable external magnetic field. RESULTS We found that magnetofection with ChABC/PEI-SPIONs significantly up-regulated the expression of ChABC in SCs. Under the driven effect of the directional magnetic field (MF), the migration of magnetofected SCs was enhanced in the direction of the magnetic force. The number of SCs with ChABC/PEI-SPIONs migrated and the distance of migration into the astrocyte region was significantly increased. The number of SCs with ChABC/PEI-SPIONs that migrated into the astrocyte region was 11.6- and 4.6-fold higher than those observed for the intact control and non-MF groups, respectively. Furthermore, it was found that SCs with ChABC/PEI-SPIONs were in close contact with astrocytes and no longer formed boundaries in the presence of MF. CONCLUSION The mobility of the SCs with ChABC/PEI-SPIONs was enhanced along the axis of MF, holding the potential to promote nerve regeneration by providing a bioactive microenvironment and relieving glial obstruction to axonal regeneration in the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Jianbo Gao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Bing Xia
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Shengyou Li
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Liangliang Huang
- Department of Orthopaedics, The General Hospital of Central Theater Command of People’s Liberation Army, Wuhan, People’s Republic of China
| | - Teng Ma
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Xiaowei Shi
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Kai Luo
- Department of Orthopaedics, The 985th Hospital of the PLA Joint Logistics Support Force, Taiyuan, People’s Republic of China
| | - Yujie Yang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Laihe Zhao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Hao Zhang
- Department of Spinal Surgery, People’s Hospital of Longhua District, Shenzhen, People’s Republic of China
| | - Beier Luo
- Department of Orthopaedics, Changhai Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| |
Collapse
|
36
|
Guevara ML, Persano F, Persano S. Nano-immunotherapy: Overcoming tumour immune evasion. Semin Cancer Biol 2019; 69:238-248. [PMID: 31883449 DOI: 10.1016/j.semcancer.2019.11.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022]
Abstract
Immunotherapy is emerging as a groundbreaking cancer treatment, offering the unprecedented opportunity to effectively treat and in several cases, even cure previously untreatable malignancies. Anti-tumour immunotherapies designed to amplify T cell responses against defined tumour antigens have long been considered effective approaches for cancer treatment. Despite a clear rationale behind such immunotherapies, extensive past efforts were unsuccessful in mediating clinically relevant anti-tumour activity in humans. This is mainly because tumours adopt specific mechanisms to circumvent the host´s immunity. Emerging data suggest that the full potential of cancer immunotherapy will be only achieved by combining immunotherapies designed to generate or amplify anti-tumour T cell responses with strategies able to impair key tumour immune-evasion mechanisms. However, many approaches aimed to re-shape the tumour immune microenvironment (TIME) are commonly associated with severe systemic toxicity, require frequent administration, and only show modest efficacy in clinical settings. The use of nanodelivery systems is revealing as a valid means to overcome these limitations by improving the targeting efficiency, minimising systemic exposure of immunomodulatory agents, and enabling the development of novel combinatorial immunotherapies. In this review, we examine the emerging field of therapeutic modulation of TIME by the use of nanoparticle-based immunomodulators and potential future directions for TIME-targeting nanotherapies.
Collapse
Affiliation(s)
- Maria L Guevara
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Francesca Persano
- Department of Mathematics and Physics, University of Salento, Lecce, Italy
| | - Stefano Persano
- Formulation Testing & Discovery, BioNTech RNA Pharmaceuticals GmbH, Mainz, Germany.
| |
Collapse
|
37
|
Kapitanova KS, Naumenko VA, Garanina AS, Melnikov PA, Abakumov MA, Alieva IB. Advances and Challenges of Nanoparticle-Based Macrophage Reprogramming for Cancer Immunotherapy. BIOCHEMISTRY (MOSCOW) 2019; 84:729-745. [PMID: 31509725 DOI: 10.1134/s0006297919070058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Despite the progress of modern medicine, oncological diseases are still among the most common causes of death of adult populations in developed countries. The current therapeutic approaches are imperfect, and the high mortality of oncological patients under treatment, the lack of personalized strategies, and severe side effects arising as a result of treatment force seeking new approaches to therapy of malignant tumors. During the last decade, cancer immunotherapy, an approach that relies on activation of the host antitumor immune response, has been actively developing. Cancer immunotherapy is the most promising trend in contemporary fundamental and practical oncology, and restoration of the pathologically altered tumor microenvironment is one of its key tasks, in particular, the reprogramming of tumor macrophages from the immunosuppressive M2-phenotype into the proinflammatory M1-phenotype is pivotal for eliciting antitumor response. This review describes the current knowledge about macrophage classification, mechanisms of their polarization, their role in formation of the tumor microenvironment, and strategies for changing the functional activity of M2-macrophages, as well as problems of targeted delivery of immunostimulatory signals to tumor macrophages using nanoparticles.
Collapse
Affiliation(s)
- K S Kapitanova
- Lomonosov Moscow State University, Department of Bioengineering and Bioinformatics, Moscow, 119991, Russia
| | - V A Naumenko
- National University of Science and Technology "MISIS", Moscow, 119049, Russia.
| | - A S Garanina
- National University of Science and Technology "MISIS", Moscow, 119049, Russia
| | - P A Melnikov
- Serbsky Federal Medical Research Center of Psychiatry and Narcology, Department of Fundamental and Applied Neurobiology, Ministry of Health of the Russian Federation, Moscow, 119034, Russia
| | - M A Abakumov
- National University of Science and Technology "MISIS", Moscow, 119049, Russia.,Russian National Research Medical University, Department of Medical Nanobiotechnology, Moscow, 117997, Russia
| | - I B Alieva
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
38
|
Han S, Wang W, Wang S, Wang S, Ju R, Pan Z, Yang T, Zhang G, Wang H, Wang L. Multifunctional biomimetic nanoparticles loading baicalin for polarizing tumor-associated macrophages. NANOSCALE 2019; 11:20206-20220. [PMID: 31621735 DOI: 10.1039/c9nr03353j] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Immunosuppression and immune tolerance lead tumor cells to evade immune system surveillance and weaken drug efficacy. The presence of various immunosuppressive cells in the tumor microenvironment, especially tumor-associated macrophages (TAMs), has been shown to be a driving force in tumor initiation and development. Reversion of the TAM phenotype is an effective way to induce a subsequent antitumor immune response. In this study, we developed baicalin-loaded poly(d,l-lactide-co-glycolide) (PLGA) nanoparticles containing an antigenic peptide (Hgp 10025-33, Hgp) and a toll-like receptor 9 agonist (CpG). The nanoparticles were further coated with a galactose-inserted erythrocyte membrane, which actively targeted the TAMs. The TAM polarization and tumor treatment effectiveness of the nanoparticles were evaluated. The biomimetic nanoparticles showed enhanced cell uptake in vitro and targeted effects in vivo. In addition, compared with baicalin-loaded PLGA-NPs (B@NPs), the biomimetic nanoparticles, such as Hgp/B@NPs-CpG and NPs@RBC-Gala, significantly polarized the TAMs such that they changed from the M2 type to the M1 type both in vitro and in vivo. Subsequently, the infiltration of CD4+ T and CD8+ T cells into tumor sites after being induced by the biomimetic nanoparticles was greatly increased, which suggested a significant enhancement of the immune activation effect and T cell response. In addition, the activation of the T cells and induction of the CTL responses effectively suppressed melanoma tumor growth in vivo. In conclusion, the biomimetic nanoparticles effectively reversed the TAM phenotype from M2 to M1, which further improved the tumor immune microenvironment and promoted tumor immunotherapy. These results suggested that the TAM-targeted biomimetic drug delivery system had the potential to reverse the phenotypes of TAMs contributing to reverse the immunosuppressive tumor microenvironment and promote tumor treatment.
Collapse
Affiliation(s)
- Shulan Han
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, P. R. China. and Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China.
| | - Wenjie Wang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, P. R. China.
| | - Shengfang Wang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, P. R. China.
| | - Shuo Wang
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China.
| | - Ruijun Ju
- Beijing Institute of Petrochemical Technology, Beijing 102617, P.R. China
| | - Zihao Pan
- Beijing Institute of Petrochemical Technology, Beijing 102617, P.R. China
| | - Tingyuan Yang
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China.
| | - Guifeng Zhang
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China.
| | - Huimei Wang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, P. R. China.
| | - Lianyan Wang
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China.
| |
Collapse
|
39
|
Abstract
Certain genetic mutations lead to the development of cancer through unchecked cell growth and division. Cancer is typically treated through surgical resection, radiotherapy, and small-molecule chemotherapy. A relatively recent approach to cancer therapy involves the use of a natural process wherein small RNA molecules regulate gene expression in a pathway known as RNA interference (RNAi). RNA oligomers pair with a network of proteins to form an RNA-induced silencing complex, which inhibits the translation of mRNA into proteins, thereby controlling the expression of gene products. Synthetically produced RNA oligomers may be designed to target and silence specific oncogenes to provide cancer therapy. The primary challenges facing the use of the RNAi pathway for cancer therapy are the safe and efficacious delivery of RNA payloads and their release at pertinent sites within disease-causing cells. Nucleases are abundant in the bloodstream and intracellular environment, and therapeutic RNA sequences often require a suitable carrier to provide protection from degradation prior to reaching their site of action in the body. The use of metal core nanoparticles (NPs) serving as targeted delivery vehicles able to shield and direct RNA payloads to their intended destinations have recently gained favor. Biological barriers present in the body establish a size prerequisite for drug delivery vehicles; to overcome recognition by the body's immune system and to gain access to intracellular environments, drug carriers must be small (< 100 nm). Iron oxide and gold core NPs can be synthesized with a high degree of control to create uniform ultrasmall drug delivery vehicles capable of bypassing key biological barriers. While progress is being made in size control of liposomal and polymer NPs, such advances still lag in comparison to the exquisite tunability and time stability of size engineering achievable with metal core NPs at bulk scales. Further, unlike lipid- and viral-based transfection agents, the biodistribution of metal core NPs can be traced using noninvasive imaging techniques that capitalize on the interaction of electromagnetic radiation and the inorganic atoms at the core of the NPs. Finally, metal core NPs have been shown to match the transfection efficiency of conventional RNA-delivery vehicles while also providing less immunogenicity and minimal side effects through the addition of tumor-targeting ligands on their surface. This Account reviews recent advances in the use of iron oxide and gold NPs for RNAi therapy. An overview of the different types of RNA-based therapies is provided along with a discussion of the advantages and current limitations of the technique. We highlight design considerations for the use of iron oxide and gold NP carriers in RNAi, including a discussion of the importance of size and its role in traversing biological barriers, NP surface modifications required for targeted delivery and RNA payload release, and auxiliary properties supporting imaging functionality for treatment monitoring. Applications of NPs for combination therapies including the pairing of RNAi with chemotherapy, photothermal therapy, immunotherapy, and radiotherapy are explored through examples. Finally, future perspectives are provided with a focus on the current limitations and the potential for clinical translation of iron oxide and gold NPs in RNAi therapy.
Collapse
Affiliation(s)
- Richard A. Revia
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Zachary R. Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|