1
|
Karati D, Meur S, Das S, Adak A, Mukherjee S. Peptide-based drugs in immunotherapy: current advances and future prospects. Med Oncol 2025; 42:177. [PMID: 40266466 DOI: 10.1007/s12032-025-02739-9] [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: 02/13/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
In immunotherapy, peptide-based medications are showing great promise as a new class of therapies that can be used to treat autoimmune diseases, cancer, and other immune-related conditions. Peptides are being created for use in immunotherapy as vaccines, immunological modulators, and adjuvants because of their capacity to precisely alter immune responses. They can imitate endogenous signals or interact with immune cells, improving the body's capacity to identify and combat malignancies or reestablishing immunological tolerance in autoimmune disorders. Notably, peptide-based treatments have demonstrated promise in promoting tumor-specific immune responses and improving the effectiveness of already available immunotherapies, such as immune checkpoint inhibitors. Notwithstanding its potential, peptide-based medications' clinical translation is fraught with difficulties, such as those pertaining to immunogenicity, bioavailability, and peptide stability. Overcoming these obstacles has been made possible by developments in peptide engineering, including pharmacokinetic optimization, receptor-binding affinity enhancement, and the creation of innovative delivery systems. The targeted distribution and effectiveness of peptide medications can be improved by using liposomes, nanoparticles, and other delivery methods, increasing their therapeutic utility. With an emphasis on recent scientific developments, mechanisms of action, and therapeutic uses, this review examines the present status of peptide-based medications in immunotherapy. We also look at the obstacles that still need to be overcome in order to get peptide-based treatments from the lab to the clinic and offer suggestions for future research initiatives. By tackling these important problems, we hope to demonstrate how peptide-based medications have the ability to revolutionize immunotherapeutic treatment approaches.
Collapse
Affiliation(s)
- Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University-TIU, Kolkata, West Bengal, 700091, India
| | - Shreyasi Meur
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, Kolkata, West Bengal, 700053, India
| | - Soumi Das
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Arpan Adak
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, Kolkata, West Bengal, 700053, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
2
|
Elkady OA, Zaafan MA, George M, Elsayed NA, Mettias VG, Edward VS, Ghataty DS. Metformin-loaded bioinspired mesoporous silica nanoparticles for targeted melanoma therapy: Nanotopographical design with in vitro and in vivo evaluation. Int J Pharm 2025; 674:125499. [PMID: 40132769 DOI: 10.1016/j.ijpharm.2025.125499] [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: 12/30/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Bioinspired nanotopographical carriers have emerged as innovative cancer therapy strategies, mimicking natural processes to enhance targeted delivery and reduce systemic toxicity. This study presents the development of virus-like mesoporous silica nanoparticles (MSN) as a delivery platform for repurposed metformin (MTF) in a topical multi-stimuli responsive system for melanoma treatment. Metformin-loaded virus-like MSN (MTF-MSN) were fabricated and incorporated into a thermo-responsive gelling system. The particles were evaluated for morphology, zeta potential (ZP), particle size (PS), entrapment efficiency (EE%), Fourier-transform infrared (FT-IR) spectroscopy, MTT cytotoxicity assay, in vitro release, and in a melanoma in vivo model. The particles exhibited a spherical morphology, a zeta potential of +31.9 ± 1.45 mV, and a particle size of 197 ± 3.47 nm, ideal for skin penetration. MTF-MSN demonstrated significant antiproliferative activity against melanoma A375 cells, with lower IC50 values (192 μg/mL) compared to free MTF (>300 μg/mL). Sustained, pH-sensitive MTF release was observed over 48 h at pH 7.4 and 6 h at pH 5.5. In vivo studies showed enhanced anti-cancer efficacy of MTF-MSN, evidenced by elevated caspase-3 and Neurofibromin Type-1 (NF-1) levels, along with suppressed angiogenesis markers VEGF and NRAS. The MTF-MSN-treated group exhibited superior outcomes compared to free MTF and controls (p < 0.05). This innovative bioinspired MTF-MSN hydrogel system optimizes MTF delivery for melanoma therapy, pioneering advancements in drug repurposing and nano-oncology.
Collapse
Affiliation(s)
- Omar A Elkady
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza 12451, Egypt
| | - Mai A Zaafan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza 12451, Egypt
| | - Marian George
- Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza 12451, Egypt
| | - Nadeen Ashraf Elsayed
- Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza 12451, Egypt
| | - Verina Ghaly Mettias
- Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza 12451, Egypt
| | - Verina Sameh Edward
- Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza 12451, Egypt
| | - Dina Saeed Ghataty
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza 12451, Egypt.
| |
Collapse
|
3
|
Mamidi N, Franco De Silva F, Orash Mahmoudsalehi A. Advanced disease therapeutics using engineered living drug delivery systems. NANOSCALE 2025; 17:7673-7696. [PMID: 40040419 DOI: 10.1039/d4nr05298f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Biological barriers significantly impede the delivery of nanotherapeutics to diseased tissues, diminishing therapeutic efficacy across pathologies such as cancer and inflammatory disorders. Although conventional strategies integrate multifunctional designs and molecular components into nanomaterials (NMs), many approaches remain insufficient to overcome these barriers. Key challenges, including inadequate drug accumulation at target sites and nonspecific biodistribution, persist in nanotherapeutic development. NMs, which harness the ability to precisely modulate drug delivery spatiotemporally and control release kinetics, represent a transformative platform for targeted cancer therapy. In this review, we highlight the biological obstacles limiting effective cancer treatment and evaluate how stimuli-responsive NMs address these constraints. By leveraging exogenous and endogenous stimuli, such NMs improve therapeutic specificity, reduce off-target effects, and amplify drug activity within pathological microenvironments. We systematically analyze the rational design and synthesis of stimuli-responsive NMs, driven by advances in oncology, biomaterials science, and nanoscale engineering. Furthermore, we highlight advances across NM classes-including polymeric, lipid-based, inorganic, and hybrid systems and explore functionalization approaches using targeting ligands, antibodies, and biomimetic coatings. Diverse delivery strategies are evaluated, such as small-molecule prodrug activation, peptide- and protein-based targeting, nucleic acid payloads, and engineered cell-mediated transport. Despite the promise of stimuli-responsive NMs, challenges such as biocompatibility, scalable fabrication, and clinical translation barriers must be addressed. By elucidating structure-function relationships and refining stimulus-triggered mechanisms, these NMs pave the way for transformative precision oncology strategies, enabling patient-specific therapies with enhanced efficacy and safety. This synthesis of interdisciplinary insights aims to catalyze innovation in next-generation nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Wisconsin Center for Nanobiosystems, School of Pharmacy, University of Wisconsin-Madison, Wisconsin-53705, USA.
| | - Fátima Franco De Silva
- Department of Food Engineering, Tecnologico de Monterrey, Monterrey, Nuevo Leon-64849, Mexico
| | - Amin Orash Mahmoudsalehi
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo Leon-64849, Mexico
| |
Collapse
|
4
|
Zhang G, Zhang X, Yue K, Zhong W. Mechanistic study of enhanced drug release in mixed pH-responsive peptide-loaded liposomes. J Biomol Struct Dyn 2025:1-15. [PMID: 40126078 DOI: 10.1080/07391102.2025.2481581] [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: 12/04/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
Liposomes serving as nanocarriers offer significant advantages in drug delivery for tumor treatment. There still exists challenges in controlling drug release by disintegrating the liposome membrane for the improvement of therapeutic efficiency. In this paper, a novel method involving the mixture of short peptides with pH-responsive characteristics into the cargo has been introduced. This approach facilitates the release of doxorubicin (DOX) in the acidic tumor tissue environment. The efficacy of this improvement was elucidated through molecular dynamics simulations and experiments. Liposomes incorporating a 1:1 ratio of peptides-DOX exhibited pronounced pH sensitivity and an enhanced drug release profile. The underlying mechanism is attributed to the peptides entering tumor tissues and undergoing protonation in acidic conditions, which increases the hydrophilicity of the peptide-DOX clusters and the internal surface tension of the liposomes. This alteration disrupts the balance between the inner and outer surface tensions of the nanocarrier, causing the liposomes to structurally disintegrate and thus enhancing drug release. The results from both thermodynamic analysis results and experimental data confirm the augmented drug release efficiency of this method, offering valuable theoretical insights for nanoparticle design and determining the optimal mixing ratio for therapeutic applications.
Collapse
Affiliation(s)
- Genpei Zhang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing, China
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| | - Xilong Zhang
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| | - Kai Yue
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing, China
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| | - Weishen Zhong
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing, China
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| |
Collapse
|
5
|
Yang K, Zhang C, Wang Z, Huang Q, Qian J, Shi G, Sun W, Wang J, Ji Y, Sun Z, Song Y, Han X. CRISPR-dCas9-Mediated PTEN Activation via Tumor Cell Membrane-Coated Nanoplatform Enhances Sensitivity to Tyrosine Kinase Inhibitors in Nonsmall Cell Lung Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13605-13616. [PMID: 39980205 DOI: 10.1021/acsami.4c21740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
EGFR tyrosine kinase inhibitors (EGFR-TKIs) have garnered substantial clinical success in treating nonsmall cell lung cancer (NSCLC) harboring epidermal growth factor receptor (EGFR) mutations. However, the inevitable emergence of drug resistance, frequently attributed to activation, mutation, or deletion of multiple signaling pathways, poses a significant challenge. Notably, the loss of PTEN protein expression has emerged as a pivotal mechanism fostering resistance in EGFR mutant lung cancers. Consequently, strategies aimed at upregulating PTEN expression hold great promise for restoring drug sensitivity. Leveraging the versatility, precision, and efficacy of nuclease-deactivated Cas9 (dCas9) as a transcriptional activator, we designed a CRISPR-dCas9 system to stimulate PTEN expression. To further enhance target specificity and drug delivery efficiency, we innovatively harnessed the tumor cell membrane (CCM) as a homologous targeting surface coating for our vector, thereby creating a targeted activation nanoplatform. Comprehensive in vitro and in vivo evaluations demonstrated that the synergistic interplay between gefitinib and the CRISPR-dCas9 system significantly enhanced drug sensitivity. The finding underscores the potential of our approach in addressing the issue of lung cancer resistance, offering a promising avenue for personalized and effective cancer therapies.
Collapse
Affiliation(s)
- Kaiyong Yang
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chunli Zhang
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zeyu Wang
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qiqing Huang
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jing Qian
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Gaoyu Shi
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wenwen Sun
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinqiu Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Ji
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhaorui Sun
- Department of Emergency Medicine, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210002, China
| | - Yanni Song
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150081, China
| | - Xin Han
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
6
|
Lorberg RY, Nair Sailaja ST, Terlau F, Victoria Cappellari M, Schmiedtchen M, Galstyan A, Strassert CA, Giese M, Voskuhl J. Photoresponsive Luminescent Silica Nanoparticles as Additive for 3D Printing and Electrospinning. Chem Asian J 2025; 20:e202401415. [PMID: 39611720 PMCID: PMC11826982 DOI: 10.1002/asia.202401415] [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: 10/14/2024] [Revised: 11/11/2024] [Accepted: 11/27/2024] [Indexed: 11/30/2024]
Abstract
In this study, we present the synthesis and a versatile way to incorporate photoresponsive organic luminophores into polymeric materials using mesoporous silica nanoparticles (MSNs). The encapsulated thioethers within the MSNs were employed in polyvinyl alcohol (PVA) films, resin-based stereolithography, and electrospinning. Due to light-induced cyclisation to dibenzothiophenes (DBTs), mmOC12 loaded materials were used to inscribe images using UV light. The DBTs formed from mmOC12 (mmDBTA/B) exhibit a long phosphorescence afterglow, which was investigated by steady-state and time-resolved photoluminescence spectroscopy. In addition, scanning electron microscopy (SEM) imaging, including energy dispersive X-ray spectroscopy (EDX), revealed the well-dispersed and intact MSNs in the polymeric materials. This approach shows a general way to incorporate non-polar organic luminophores into polymeric materials while retaining their unique emission properties.
Collapse
Affiliation(s)
- Rick Y. Lorberg
- Faculty of Chemistry (Organic Chemistry)Center of Medical Biotechnology (ZMB)Center for Nanointegration Duisburg-Essen (CENIDE)University of Duisburg-EssenUniversitätsstraße 7Essen45117Germany
| | - Sidharth Thulaseedharan Nair Sailaja
- Faculty of Chemistry (Organic Chemistry)Center of Medical Biotechnology (ZMB)Center for Nanointegration Duisburg-Essen (CENIDE)University of Duisburg-EssenUniversitätsstraße 7Essen45117Germany
| | - Fabian Terlau
- Faculty of ChemistryCenter for Nanointegration Duisburg-Essen (CENIDE), Center of Medical Biotechnology (ZMB) and Center for Water and Environment Research (ZWU)University of Duisburg-EssenUniversitätsstraße 5Essen45141Germany
| | - Maria Victoria Cappellari
- Institute for Inorganic and Analytical ChemistryUniversity of MünsterCorrensstraße 28/30Münster48149Germany
- CiMIC, SoN, CeNTechUniversity of MünsterHeisenbergstraße 11Münster48149Germany
| | - Marco Schmiedtchen
- Faculty of Chemistry (Organic Chemistry)Center of Medical Biotechnology (ZMB)Center for Nanointegration Duisburg-Essen (CENIDE)University of Duisburg-EssenUniversitätsstraße 7Essen45117Germany
| | - Anzhela Galstyan
- Faculty of ChemistryCenter for Nanointegration Duisburg-Essen (CENIDE), Center of Medical Biotechnology (ZMB) and Center for Water and Environment Research (ZWU)University of Duisburg-EssenUniversitätsstraße 5Essen45141Germany
| | - Cristian A. Strassert
- Institute for Inorganic and Analytical ChemistryUniversity of MünsterCorrensstraße 28/30Münster48149Germany
- CiMIC, SoN, CeNTechUniversity of MünsterHeisenbergstraße 11Münster48149Germany
| | - Michael Giese
- Faculty of Chemistry (Organic Chemistry)Center of Medical Biotechnology (ZMB)Center for Nanointegration Duisburg-Essen (CENIDE)University of Duisburg-EssenUniversitätsstraße 7Essen45117Germany
- GUIDE Co-Creation Lab Produktinnovationen (CCLP)Schützenbahn 70Essen45127Germany
| | - Jens Voskuhl
- Faculty of Chemistry (Organic Chemistry)Center of Medical Biotechnology (ZMB)Center for Nanointegration Duisburg-Essen (CENIDE)University of Duisburg-EssenUniversitätsstraße 7Essen45117Germany
| |
Collapse
|
7
|
Yousefiasl S, Ghovvati M, Alibakhshi A, Azizi M, Samadi P, Kumar A, Shojaeian A, Sharifi E, Zare EN, Dey AD, Chehelgerdi M, Makvandi P. Smart Mesoporous Silica Nanoparticles in Cancer: Diagnosis, Treatment, Immunogenicity, and Clinical Translation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408898. [PMID: 39840493 DOI: 10.1002/smll.202408898] [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: 09/28/2024] [Revised: 01/01/2025] [Indexed: 01/23/2025]
Abstract
In cancer research and personalized medicine, mesoporous silica nanoparticles (MSNs) have emerged as a significant breakthrough in both cancer treatment and diagnosis. MSNs offer targeted drug delivery, enhancing therapeutic effectiveness while minimizing adverse effects on healthy cells. Due to their unique characteristics, MSNs provide targeted drug delivery, maximizing therapeutic effectiveness with minimal adverse effects on healthy cells. The review thoroughly investigates the role of MSNs as potent drug carriers, noted for their high drug-loading capacity and controlled release, which significantly improves drug permeability and retention. Additionally, it discusses surface modification techniques that enable MSNs to target cancer cells precisely. The manuscript provides comprehensive insights into various MSN applications, including their role in cancer diagnosis, the design of advanced biosensors, and the development of both conventional and stimuli-responsive drug delivery platforms. Special focus is given to stimuli-triggered MSN systems, responsive to internal stimuli (e.g., pH, redox, enzyme) and external stimuli (e.g., temperature, magnetic field, light, ultrasound), highlighting the cutting-edge progress in MSN technology. Additionally, the review delves into the immunogenicity and biosafety aspects of MSNs, underscoring their potential for clinical translation. Besides summarizing the current state of MSN research in oncology, this review also illuminates the path for future advancements and clinical applications.
Collapse
Affiliation(s)
- Satar Yousefiasl
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Mahsa Ghovvati
- Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA, 90095, USA
| | - Abbas Alibakhshi
- Cancer Research Center, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, 6517838636, Iran
| | - Mehdi Azizi
- Cancer Research Center, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, 6517838636, Iran
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | - Pouria Samadi
- Poursina Hakim Digestive Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, 8198314271, Iran
| | - Arun Kumar
- Department of Pharmacy, School of Health Sciences, Central University of South Bihar, Gaya, 824209, India
| | - Ali Shojaeian
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, 6517838636, Iran
| | - Esmaeel Sharifi
- Cancer Research Center, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, 6517838636, Iran
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | | | - Asmita Deka Dey
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
- University Centre for Research & Development, Chandigarh University, Mohali, Punjab, 140413, India
- Chitkara Centre for Research and Development, Chitkara University, Himachal Pradesh, 174103, India
| |
Collapse
|
8
|
Wu X, Ma L, Zhang Y, Liu S, Cheng L, You C, Dong Z. Application progress of nanomaterials in the treatment of prostate cancer. ANNALES PHARMACEUTIQUES FRANÇAISES 2025; 83:1-12. [PMID: 39187009 DOI: 10.1016/j.pharma.2024.08.009] [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: 07/12/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
Prostate cancer is one of the most common malignant tumors in men, which seriously threatens the survival and quality of life of patients. At present, there are serious limitations in the treatment of prostate cancer, such as drug tolerance, drug resistance and easy recurrence. Sonodynamic therapy and chemodynamic therapy are two emerging tumor treatment methods, which activate specific drugs or sonosensitizers through sound waves or chemicals to produce reactive oxygen species and kill tumor cells. Nanomaterials are a kind of nanoscale materials with many excellent physical properties such as high targeting, drug release regulation and therapeutic monitoring. Sonodynamic therapy and chemodynamic therapy combined with the application of nanomaterials can improve the therapeutic effect of prostate cancer, reduce side effects and enhance tumor immune response. This article reviews the application progress of nanomaterials in the treatment of prostate cancer, especially the mechanism, advantages and challenges of nanomaterials in sonodynamic therapy and chemodynamic therapy, which provides new ideas and prospects for research in this field.
Collapse
Affiliation(s)
- Xuewu Wu
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Longtu Ma
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Yang Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, China
| | - Shuai Liu
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Long Cheng
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Chengyu You
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Zhilong Dong
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China.
| |
Collapse
|
9
|
Leng Y, Wu Y, Xiao W, Su X, Liu Z. Dynamic-Covalent Mesoporous Silica Nanohybrid with pH/ROS-Responsive Drug Release for Targeted Tumor Therapy. ACS OMEGA 2024; 9:47428-47435. [PMID: 39651082 PMCID: PMC11618396 DOI: 10.1021/acsomega.4c04502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 10/27/2024] [Accepted: 11/07/2024] [Indexed: 12/11/2024]
Abstract
Nanomedicine provides promising new methodologies for the treatment of tumors but still faces several limitations, including poor colloidal stability, uncontrollable drug release, and insufficient drug targeting. Herein, hyaluronic acid (HA) was used to modify the surface of mesoporous silica nanoparticles (MSNs) via a dynamic-covalent linker, phenylborate ester (PBAE), termed MA. The HA modifier provided enhanced colloidal stability to the hybrid nanoparticles. As expected, MA exhibited an improved biocompatibility and high potential for biomedical applications. Moreover, MA with a negatively charged surface effectively adsorbed the drug Doxorubicin (DOX) inside the carriers, ensuring minimal drug leakage. In an acidic and reactive oxygen species (ROS)-containing condition mimicking the tumor microenvironment, MA@DOX (MAD) continuously released its payloads, likely due to the cleavage of the pH/ROS-sensitive PBAE. Compared with free DOX, MAD had 2.2 times higher accessibility to tumor cells than free DOX. The favorable stability and cancer-selective drug release make this nanoformulation a promising platform for potent cancer treatment.
Collapse
Affiliation(s)
- Yurong Leng
- School
of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- Jiangxi
Province Key Laboratory of Oral Biomedicine, Nanchang 330006, China
- Jiangxi
Province Clinical Research Center for Oral Diseases, Nanchang 330006, China
| | - Yanmei Wu
- School
of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- Jiangxi
Province Key Laboratory of Oral Biomedicine, Nanchang 330006, China
- Jiangxi
Province Clinical Research Center for Oral Diseases, Nanchang 330006, China
| | - Wenjing Xiao
- School
of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Xiaoquan Su
- School
of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Zhe Liu
- School
of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- Jiangxi
Province Key Laboratory of Oral Biomedicine, Nanchang 330006, China
- Jiangxi
Province Clinical Research Center for Oral Diseases, Nanchang 330006, China
| |
Collapse
|
10
|
Yan J, Siwakoti P, Shaw S, Bose S, Kokil G, Kumeria T. Porous silicon and silica carriers for delivery of peptide therapeutics. Drug Deliv Transl Res 2024; 14:3549-3567. [PMID: 38819767 PMCID: PMC11499345 DOI: 10.1007/s13346-024-01609-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2024] [Indexed: 06/01/2024]
Abstract
Peptides have gained tremendous popularity as biological therapeutic agents in recent years due to their favourable specificity, diversity of targets, well-established screening methods, ease of production, and lower cost. However, their poor physiological and storage stability, pharmacokinetics, and fast clearance have limited their clinical translation. Novel nanocarrier-based strategies have shown promise in overcoming these issues. In this direction, porous silicon (pSi) and mesoporous silica nanoparticles (MSNs) have been widely explored as potential carriers for the delivery of peptide therapeutics. These materials possess several advantages, including large surface areas, tunable pore sizes, and adjustable pore architectures, which make them attractive carriers for peptide delivery systems. In this review, we cover pSi and MSNs as drug carriers focusing on their use in peptide delivery. The review provides a brief overview of their fabrication, surface modification, and interesting properties that make them ideal peptide drug carriers. The review provides a systematic account of various studies that have utilised these unique porous carriers for peptide delivery describing significant in vitro and in vivo results. We have also provided a critical comparison of the two carriers in terms of their physicochemical properties and short-term and long-term biocompatibility. Lastly, we have concluded the review with our opinion of this field and identified key areas for future research for clinical translation of pSi and MSN-based peptide therapeutic formulations.
Collapse
Affiliation(s)
- Jiachen Yan
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Prakriti Siwakoti
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Siuli Shaw
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, 201301, India
| | - Sudeep Bose
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, 201301, India
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, 201301, India
| | - Ganesh Kokil
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia.
- Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Tushar Kumeria
- School of Materials Science and Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia.
- Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW, 2052, Australia.
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
11
|
Grayton QE, El-Ahmad H, Lynch AL, Nogler ME, Wallet SM, Schoenfisch MH. Nitric Oxide-Releasing Topical Treatments for Cutaneous Melanoma. Mol Pharm 2024; 21:5632-5645. [PMID: 39353049 PMCID: PMC11875128 DOI: 10.1021/acs.molpharmaceut.4c00618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Melanoma is an aggressive skin cancer notorious for high levels of drug resistance. Additionally, current treatments such as immunotherapies are often associated with numerous adverse side effects. The use of nitric oxide (NO) may represent an attractive treatment for melanoma due to NO's various anticancer properties, unlikeliness to foster resistance, and limited toxicity toward healthy tissues. The anticancer effects of chemical NO donors have been explored previously but with limited understanding of the needed characteristics for exerting optimal antimelanoma activity. Herein, the in vitro therapeutic efficacy of three macromolecular NO donor systems (i.e., cyclodextrin, mesoporous silica nanoparticles, and hyaluronic acid) with tunable NO-release kinetics was explored by evaluating skin permeation along with toxicity against melanoma and healthy skin cells. Cytotoxicity against melanoma cells was dependent on NO payload and not donor identity or NO-release kinetics. In contrast, cytotoxicity against healthy cells was primarily influenced by the macromolecular NO donor, with cyclodextrin- and hyaluronic acid-based NO donors having the highest therapeutic indices. In vitro skin permeation was influenced by both the size and charge of the NO donor, with smaller, more neutral donors resulting in greater permeation. A Pluronic F127 organogel was optimized for the delivery of a cyclodextrin-based NO donor. Delivery of the NO donor in this manner resulted in increased in vitro skin permeation and reduced tumor growth in an in vivo model.
Collapse
Affiliation(s)
- Quincy E Grayton
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Heba El-Ahmad
- Department of Oral Biology, University of Florida, Gainesville, Florida 32610, United States
| | - Anna L Lynch
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mikaylin E Nogler
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Shannon M Wallet
- Department of Oral Biology, University of Florida, Gainesville, Florida 32610, United States
| | - Mark H Schoenfisch
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
12
|
Tu B, Jonnalagadda S. Amorphous stabilization of BCS II drugs using mesoporous silica. Int J Pharm 2024; 663:124555. [PMID: 39111354 DOI: 10.1016/j.ijpharm.2024.124555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
This study aimed to investigate the amorphous stabilization of BCS Class II drugs using mesoporous silica as a carrier to produce amorphous solid dispersions. Ibuprofen, fenofibrate, and budesonide were selected as model drugs to evaluate the impact of molecular weight and partition coefficient on the solid state of drug-loaded mesoporous silica (MS) particles. The model drugs were loaded into three grades of MS, SYLYSIA SY730, SYLYSIA SY430, and SYLYSIA SY350, with pore diameters of 2.5 nm, 17 nm, and 21 nm, respectively, at 1:1, 2:1, and 3:1, carrier to drug ratios, and three different loading concentrations using solvent immersion and spray drying techniques. Differential scanning calorimetry (DSC) thermograms of SY430 and SY350 samples exhibited melting point depressions indicating constricted crystallization inside the pores, whereas SY730 samples with melting points matching the pure API may be a result of surface crystallization. Powder x-ray diffraction (PXRD) diffractograms showed all crystalline samples matched the diffraction patterns of the pure API indicating no polymorphic transitions and all 3:1 ratio samples exhibited amorphous halo profiles. Response surface regression analysis and Classification and Regression Tree (CART) analysis suggest carrier to drug ratios, followed by molecular weight, have the most significant impact on the crystallinity of a drug loaded into MS particles.
Collapse
Affiliation(s)
- Buu Tu
- Saint Joseph's University, 600 S 43rd Street, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
13
|
Jeon Y, Kim TR, Park ES, Park JH, Youn HS, Hwang DY, Seo S. Effect of Silica Nanoparticle Treatment on Adhesion between Tissue-like Substrates and In Vivo Skin Wound Sealing. J Funct Biomater 2024; 15:259. [PMID: 39330234 PMCID: PMC11433542 DOI: 10.3390/jfb15090259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024] Open
Abstract
Silica nanoparticles are innovative solutions of surgical glue that can readily adhere to various tissue-like substrates without the need for time-consuming chemical reactions or ultraviolet irradiation. Herein, 10 nm-sized silica nanoparticle (SiNP10) treatment exhibited maximum adhesion strength in the porcine heart tissue model, which was approximately 7.15 times higher than that of the control group of non-treatment. We assessed the effects of silica nanoparticle treatment on in vivo skin wounds by scoring tissue adhesion and inflammation using histological images. Compared to the commercial cyanoacrylate skin adhesive (Dermabond), suppression of inflammatory cytokine levels in the incision wound skin was observed. We further quantified the expression of angiogenic growth factors and connective tissue formation-related proteins. On day 5 after wound closing treatment, the expression levels of PDGF-BB growth factor were significantly higher in SiNP10 treatment (0.64 ± 0.03) compared to Dermabond (0.07 ± 0.05). This stimulated angiogenesis and connective tissue formation in the skin of the incision wound may be associated with the promoting effects of SiNP10 treatment on wound closure and tissue adhesion.
Collapse
Affiliation(s)
- Yeji Jeon
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Tae Ryeol Kim
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Eun Seo Park
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Jae Hyun Park
- Young Chemical Co., Ltd., 80-93, Golden root-ro, Juchon-myeon, Gimhae 50969, Republic of Korea
| | - Han Sung Youn
- Young Chemical Co., Ltd., 80-93, Golden root-ro, Juchon-myeon, Gimhae 50969, Republic of Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Sungbaek Seo
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| |
Collapse
|
14
|
Yang Y, Yang S, Zhang B, Wang J, Meng D, Cui L, Zhang L. Hybrid Liposome-MSN System with Co-Delivering Potential Effective Against Multidrug-Resistant Tumor Targets in Mice Model. Int J Nanomedicine 2024; 19:8949-8970. [PMID: 39246424 PMCID: PMC11378800 DOI: 10.2147/ijn.s472276] [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: 04/04/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction RNA interference (RNAi) stands as a widely employed gene interference technology, with small interfering RNA (siRNA) emerging as a promising tool for cancer treatment. However, the inherent limitations of siRNA, such as easy degradation and low bioavailability, hamper its efficacy in cancer therapy. To address these challenges, this study focused on the development of a nanocarrier system (HLM-N@DOX/R) capable of delivering both siRNA and doxorubicin for the treatment of breast cancer. Methods The study involved a comprehensive investigation into various characteristics of the nanocarrier, including shape, diameter, Fourier transform infrared (FT-IR) spectroscopy, X-ray photoelectron spectroscopy (XPS), encapsulation efficiency, and drug loading. Subsequently, in vitro and in vivo studies were conducted on cytotoxicity, cellular uptake, cellular immunofluorescence, lysosome escape, and mouse tumor models to evaluate the efficacy of the nanocarrier in reversing tumor multidrug resistance and anti-tumor effects. Results The results showed that HLM-N@DOX/R had a high encapsulation efficiency and drug loading capacity, and exhibited pH/redox dual responsive drug release characteristics. In vitro and in vivo studies showed that HLM-N@DOX/R inhibited the expression of P-gp by 80%, inhibited MDR tumor growth by 71% and eliminated P protein mediated multidrug resistance. Conclusion In summary, HLM-N holds tremendous potential as an effective and targeted co-delivery system for DOX and P-gp siRNA, offering a promising strategy for overcoming MDR in breast cancer.
Collapse
MESH Headings
- Animals
- Doxorubicin/pharmacology
- Doxorubicin/chemistry
- Doxorubicin/pharmacokinetics
- Doxorubicin/administration & dosage
- Female
- Liposomes/chemistry
- Mice
- Drug Resistance, Neoplasm/drug effects
- Humans
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/chemistry
- RNA, Small Interfering/pharmacokinetics
- Drug Resistance, Multiple/drug effects
- Breast Neoplasms/drug therapy
- Cell Line, Tumor
- MCF-7 Cells
- Mice, Inbred BALB C
- Drug Carriers/chemistry
- Drug Carriers/pharmacokinetics
- Nanoparticles/chemistry
- Drug Liberation
- Antibiotics, Antineoplastic/pharmacology
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/pharmacokinetics
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yanan Yang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, People's Republic of China
| | - Shuoye Yang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, People's Republic of China
- Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, People's Republic of China
| | - Beibei Zhang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, People's Republic of China
| | - Jinpeng Wang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, People's Republic of China
| | - Di Meng
- School of Biological Engineering, Henan University of Technology, Zhengzhou, People's Republic of China
| | - Lan Cui
- School of Biological Engineering, Henan University of Technology, Zhengzhou, People's Republic of China
- Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, People's Republic of China
| | - Lu Zhang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, People's Republic of China
- Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, People's Republic of China
| |
Collapse
|
15
|
Yang S, Raza F, Li K, Qiu Y, Su J, Qiu M. Maximizing arsenic trioxide's anticancer potential: Targeted nanocarriers for solid tumor therapy. Colloids Surf B Biointerfaces 2024; 241:114014. [PMID: 38850742 DOI: 10.1016/j.colsurfb.2024.114014] [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: 03/24/2024] [Revised: 05/18/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Arsenic trioxide (ATO) has gained significant attention due to its promising therapeutic effects in treating different diseases, particularly acute promyelocytic leukemia (APL). Its potent anticancer mechanisms have been extensively studied. Despite the great efficacy ATO shows in fighting cancers, drawbacks in the clinical use are obvious, especially for solid tumors, which include rapid renal clearance and short half-life, severe adverse effects, and high toxicity to normal cells. Recently, the emergence of nanomedicine offers a potential solution to these limitations. The enhanced biocompatibility, excellent targeting capability, and desirable effectiveness have attracted much interest. Therefore, we summarized various nanocarriers for targeted delivery of ATO to solid tumors. We also provided detailed anticancer mechanisms of ATO in treating cancers, its clinical trials and shortcomings as well as the combination therapy of ATO and other chemotherapeutic agents for reduced drug resistance and synergistic effects. Finally, the future study direction and prospects were also presented.
Collapse
Affiliation(s)
- Shiqi Yang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kunwei Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yujiao Qiu
- The Wharton School and School of Nursing, University of Pennsylvania, Philadelphia 19104, USA
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
16
|
Mohanan S, Guan X, Liang M, Karakoti A, Vinu A. Stimuli-Responsive Silica Silanol Conjugates: Strategic Nanoarchitectonics in Targeted Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2301113. [PMID: 36967548 DOI: 10.1002/smll.202301113] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The design of novel drug delivery systems is exceptionally critical in disease treatments. Among the existing drug delivery systems, mesoporous silica nanoparticles (MSNs) have shown profuse promise owing to their structural stability, tunable morphologies/sizes, and ability to load different payload chemistry. Significantly, the presence of surface silanol groups enables functionalization with relevant drugs, imaging, and targeting agents, promoting their utility and popularity among researchers. Stimuli-responsive silanol conjugates have been developed as a novel, more effective way to conjugate, deliver, and release therapeutic drugs on demand and precisely to the selected location. Therefore, it is urgent to summarize the current understanding and the surface silanols' role in making MSN a versatile drug delivery platform. This review provides an analytical understanding of the surface silanols, chemistry, identification methods, and their property-performance correlation. The chemistry involved in converting surface silanols to a stimuli-responsive silica delivery system by endogenous/exogenous stimuli, including pH, redox potential, temperature, and hypoxia, is discussed in depth. Different chemistries for converting surface silanols to stimuli-responsive bonds are discussed in the context of drug delivery. The critical discussion is culminated by outlining the challenges in identifying silanols' role and overcoming the limitations in synthesizing stimuli-responsive mesoporous silica-based drug delivery systems.
Collapse
Affiliation(s)
- Shan Mohanan
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Xinwei Guan
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Mingtao Liang
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, The University of Newcastle, Callaghan, 2308, Australia
| | - Ajay Karakoti
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| | - Ajayan Vinu
- Global Innovative Centre for Advanced Nanomaterials, The School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, 2308, Australia
| |
Collapse
|
17
|
Song Y, Zhang J, Zhu L, Zhang H, Wu G, Liu T. Recent advances in nanodelivery systems of resveratrol and their biomedical and food applications: a review. Food Funct 2024; 15:8629-8643. [PMID: 39140384 DOI: 10.1039/d3fo03892k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Resveratrol is a non-flavonoid polyphenolic compound with numerous functional properties, such as anticancer, anti-inflammation, anti-oxidation, anti-obesity and more. However, resveratrol's poor solubility within aqueous media and low stability usually lead to compromised bioavailability, ultimately limiting its uptake and applications. Nanodelivery technologies have been studied intensively due to their potential in effectively improving resveratrol properties, thereby providing promising solutions for enhancing the bioavailability of resveratrol. Thus, this article aimed to review the recent advances of resveratrol nanodelivery systems, specifically on the types of nanodelivery systems, the corresponding preparation principles, advantages, as well as potential limitations associated. Meanwhile, studies have also found that coupled with nanodelivery systems, the functional properties of resveratrol could trigger apoptosis in cancer cells and inflammatory cells through various signaling pathways. Therefore, this article will also lead into discussions on the application aspects of resveratrol nanodelivery systems, emphasizing toward the fields of biomedical and food sciences. Potential pitfalls of resveratrol nanodelivery systems, such as issues with toxicity and target release, as well as outlooks regarding resveratrol nanodelivery systems are included in the Conclusion section, in the hope to provide insights for relevant future research.
Collapse
Affiliation(s)
- Yanan Song
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - Junjia Zhang
- Department of Food Science, Rutgers, The State University of New Jersey, 65 Dudley Road, New Brunswick, NJ 08901, USA
| | - Ling Zhu
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - Hui Zhang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - Gangcheng Wu
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - Tongtong Liu
- Binzhou Zhongyu Food Company Limited, Key Laboratory of Wheat Processing, Ministry of Agriculture and Rural Affairs, National Industry Technical Innovation Center for Wheat Processing, Bohai Advanced Technology Institute, Binzhou 256600, China
| |
Collapse
|
18
|
Karnopp JCF, Jorge J, da Silva JR, Boldo D, Del Pino Santos KF, Duarte AP, de Castro GR, de Azevedo RB, Prada AL, Amado JRR, Martines MAU. Synthesis, Characterization, and Cytotoxicity Evaluation of Chlorambucil-Functionalized Mesoporous Silica Nanoparticles. Pharmaceutics 2024; 16:1086. [PMID: 39204431 PMCID: PMC11359805 DOI: 10.3390/pharmaceutics16081086] [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: 05/31/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
This study describes the synthesis and characterization of chlorambucil (CLB)-functionalized mesoporous silica nanoparticles (MSNs) for potential application in cancer therapy. The nanoparticles were designed with a diameter between 20 and 50 nm to optimize cellular uptake and avoid rapid clearance from the bloodstream. The synthesis method involved modifying a previously reported technique to reduce particle size. Successful functionalization with CLB was confirmed through various techniques, including Fourier transform infrared spectroscopy (FTIR) and elemental analysis. The cytotoxicity of the CLB-functionalized nanoparticles (MSN@NH2-CLB) was evaluated against human lung adenocarcinoma cells (A549) and colon carcinoma cells (CT26WT). The results suggest significantly higher cytotoxicity of MSN@NH2-CLB compared to unbound CLB, with improved selectivity towards cancer cells over normal cells. This suggests that MSN@NH2-CLB holds promise as a drug delivery system for targeted cancer therapy.
Collapse
Affiliation(s)
- Juliana Camila Fischer Karnopp
- Postgraduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, MS, Brazil; (J.C.F.K.); (J.J.); (D.B.); (K.F.D.P.S.); (A.P.D.)
| | - Juliana Jorge
- Postgraduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, MS, Brazil; (J.C.F.K.); (J.J.); (D.B.); (K.F.D.P.S.); (A.P.D.)
| | - Jaqueline Rodrigues da Silva
- Postgraduate Program in Nanoscience and Nanotechnology, Biological Science Institute, University of Brasilia, Brasilia 70910-900, DF, Brazil; (J.R.d.S.); (R.B.d.A.)
| | - Diego Boldo
- Postgraduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, MS, Brazil; (J.C.F.K.); (J.J.); (D.B.); (K.F.D.P.S.); (A.P.D.)
| | - Kristiane Fanti Del Pino Santos
- Postgraduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, MS, Brazil; (J.C.F.K.); (J.J.); (D.B.); (K.F.D.P.S.); (A.P.D.)
| | - Adriana Pereira Duarte
- Postgraduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, MS, Brazil; (J.C.F.K.); (J.J.); (D.B.); (K.F.D.P.S.); (A.P.D.)
| | - Gustavo Rocha de Castro
- Postgraduate Program in Environmental Biotechnology, Bioscience Institute, Sao Paulo State University, Botucatu 18618-000, SP, Brazil;
| | - Ricardo Bentes de Azevedo
- Postgraduate Program in Nanoscience and Nanotechnology, Biological Science Institute, University of Brasilia, Brasilia 70910-900, DF, Brazil; (J.R.d.S.); (R.B.d.A.)
| | - Ariadna Lafourcade Prada
- Postgraduate Program in Biotechnology, Faculty of Pharmacy, Food and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil;
| | - Jesús Rafael Rodríguez Amado
- Postgraduate Program in Health Sciences, Faculty of Health Sciences, Federal University of Grande Dourados, Dourados 79804-970, MS, Brazil;
| | - Marco Antonio Utrera Martines
- Postgraduate Program in Chemistry, Chemistry Institute, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, MS, Brazil; (J.C.F.K.); (J.J.); (D.B.); (K.F.D.P.S.); (A.P.D.)
| |
Collapse
|
19
|
Nair A, Chandrashekhar H R, Day CM, Garg S, Nayak Y, Shenoy PA, Nayak UY. Polymeric functionalization of mesoporous silica nanoparticles: Biomedical insights. Int J Pharm 2024; 660:124314. [PMID: 38862066 DOI: 10.1016/j.ijpharm.2024.124314] [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: 03/04/2024] [Revised: 05/25/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024]
Abstract
Mesoporous silica nanoparticles (MSNs) endowed with polymer coatings present a versatile platform, offering notable advantages such as targeted, pH-controlled, and stimuli-responsive drug delivery. Surface functionalization, particularly through amine and carboxyl modification, enhances their suitability for polymerization, thereby augmenting their versatility and applicability. This review delves into the diverse therapeutic realms benefiting from polymer-coated MSNs, including photodynamic therapy (PDT), photothermal therapy (PTT), chemotherapy, RNA delivery, wound healing, tissue engineering, food packaging, and neurodegenerative disorder treatment. The multifaceted potential of polymer-coated MSNs underscores their significance as a focal point for future research endeavors and clinical applications. A comprehensive analysis of various polymers and biopolymers, such as polydopamine, chitosan, polyethylene glycol, polycaprolactone, alginate, gelatin, albumin, and others, is conducted to elucidate their advantages, benefits, and utilization across biomedical disciplines. Furthermore, this review extends its scope beyond polymerization and biomedical applications to encompass topics such as surface functionalization, chemical modification of MSNs, recent patents in the MSN domain, and the toxicity associated with MSN polymerization. Additionally, a brief discourse on green polymers is also included in review, highlighting their potential for fostering a sustainable future.
Collapse
Affiliation(s)
- Akhil Nair
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Raghu Chandrashekhar H
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Candace M Day
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Sanjay Garg
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Padmaja A Shenoy
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Usha Y Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
20
|
Saadh MJ, Mustafa MA, Malathi H, Ahluwalia G, Kaur S, Al-Dulaimi MAAH, Alubiady MHS, Zain Al-Abdeen SH, Shakier HG, Ali MS, Ahmad I, Abosaoda MK. Targeting the pancreatic tumor microenvironment by plant-derived products and their nanoformulations. Med Oncol 2024; 41:201. [PMID: 39001987 DOI: 10.1007/s12032-024-02443-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 06/27/2024] [Indexed: 07/15/2024]
Abstract
Pancreatic cancer remains a significant health issue with limited treatment options. The tumor stroma, a complex environment made up of different cells and proteins, plays a crucial role in tumor growth and chemoresistance. Targeting tumor stroma, consisting of diverse non-tumor cells such as fibroblasts, extracellular matrix (ECM), immune cells, and also pre-vascular cells is encouraging for remodeling solid cancers, such as pancreatic cancer. Remodeling the stroma of pancreas tumors can be suggested as a strategy for reducing resistance to chemo/immunotherapy. Several studies have shown that phytochemicals from plants can affect the tumor environment and have anti-cancer properties. By targeting key pathways involved in stromal activation, phytochemicals may disrupt communication between the tumor and stroma and make tumor cells more sensitive to different treatments. Additionally, phytochemicals have immunomodulatory and anti-angiogenic properties, all of which contribute to their potential in treating pancreatic cancer. This review will provide a detailed look at how phytochemicals impact the tumor stroma and their effects on pancreatic tumor growth, spread, and response to treatment. It will also explore the potential of combining phytochemicals with other treatment options like chemotherapy, immunotherapy, and radiation.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, University of Imam Jaafar AL-Sadiq, Baghdad, Iraq
| | - H Malathi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Gunveen Ahluwalia
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, 303012, Rajasthan, India
| | - Sumeet Kaur
- Department of Applied Sciences, Chandigarh Engineering Colleges, Chandigarh Group of Colleges, Jhanjeri, 140307, Mohali, Punjab, India
| | | | | | | | | | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
21
|
Liang T, Liu X, Tong Y, Ding Q, Yang M, Ning H. Recent Advances in Targeted Therapies for Infantile Hemangiomas. Int J Nanomedicine 2024; 19:6127-6143. [PMID: 38911507 PMCID: PMC11193998 DOI: 10.2147/ijn.s463119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/02/2024] [Indexed: 06/25/2024] Open
Abstract
Targeted therapy for infantile hemangiomas (IHs) has been extensively studied as they can concentrate drugs, increase therapeutic efficacy and reduce drug dosage. Meanwhile, they can extend drug release times, enhance drug stability, decrease dosing frequency, and improve patient compliance. Moreover, carriers made from biocompatible materials reduced drug immunogenicity, minimizing adverse reactions. However, current targeted formulations still face numerous challenges such as the non-absolute safety of carrier materials; the need to further increase drug loading capacity; the limitation of animal hemangioma models in fully replicating the biological properties of human infantile hemangiomas; the establishment of models for deep-seated hemangiomas with high incidence rates; and the development of more specific targets or markers. In this review, we provided a brief overview of the characteristics of IHs and summarized the past decade's advances, advantages, and targeting strategies of targeted drug delivery systems for IHs and discussed their applications in the treatment of IHs. Furthermore, the goal is to provide a reference for further research and application in this field.
Collapse
Affiliation(s)
- Tiantian Liang
- Department of Pharmacy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Xianbin Liu
- Department of Pharmacy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Yujun Tong
- Department of Breast Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Qian Ding
- Department of Clinical Pharmacy, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan, People’s Republic of China
| | - Min Yang
- Department of Pharmacy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Hong Ning
- Department of Pharmacy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| |
Collapse
|
22
|
Grayton QE, Phan TT, Kussatz CC, Schoenfisch MH. Hyaluronic Acid-Coated Silica Nanoparticles for Targeted Delivery of Nitric Oxide to Cancer Cells. ACS APPLIED BIO MATERIALS 2024; 7:3796-3809. [PMID: 38776418 PMCID: PMC11546759 DOI: 10.1021/acsabm.4c00171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Drug resistance and off-target toxicity are two of the greatest challenges to chemotherapeutic melanoma treatments. Nitric oxide (NO) represents an attractive alternative to conventional therapeutics due to its numerous anticancer properties and low probability of engendering resistance. As NO is highly reactive, macromolecular NO donors are needed for the controlled and targeted delivery of NO for therapeutic applications. Herein, mesoporous silica nanoparticles (MSNs) coated with hyaluronic acid (HA) were developed as a NO delivery system to facilitate controlled delivery to cancer cells through both passive and active targeting via the enhanced permeation and retention effect and directed binding of HA with CD44 receptors, respectively. The aminosilane modification, HA concentration, and HA molecular weight were systematically evaluated to facilitate the MSN coating and NO loading. The hydrodynamic diameter and dispersity of the nanoparticles increased after HA coating due to the hydrophilic nature of HA, with greater increases observed at higher HA molecular weight. Lower starting concentrations of HA and aminosilanes with longer alkyl chains favored more efficient HA coating. Faster NO-release kinetics and lower NO payloads were observed for the HA-coated MSNs relative to uncoated MSNs. However, the localized delivery of NO to cancer cells through the active targeting conferred by HA increased levels of oxidative stress and induced mitochondria-mediated apoptosis in melanoma cells. Cytotoxicity was also evaluated against human dermal fibroblasts, with the use of 6 kDa HA-coated MSNs resulting in the greatest therapeutic indices. Enhanced internalization of HA-coated nanoparticles into melanoma cells versus uncoated nanoparticles was visualized with confocal microscopy and quantified by fluorescence spectroscopy. In total, HA-coated MSNs represent a promising NO delivery system for potential use as a chemotherapeutic for skin melanomas.
Collapse
Affiliation(s)
- Quincy E. Grayton
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill NC 27599
| | - Tien T. Phan
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill NC 27599
| | - Caden C. Kussatz
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill NC 27599
| | - Mark H. Schoenfisch
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill NC 27599
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill NC 27599
| |
Collapse
|
23
|
Slapak EJ, El Mandili M, Ten Brink MS, Kros A, Bijlsma MF, Spek CA. CAPN2-responsive mesoporous silica nanoparticles: A promising nanocarrier for targeted therapy of pancreatic cancer. Cancer Lett 2024; 590:216845. [PMID: 38589004 DOI: 10.1016/j.canlet.2024.216845] [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: 10/26/2023] [Revised: 02/29/2024] [Accepted: 03/28/2024] [Indexed: 04/10/2024]
Abstract
Pancreatic adenocarcinoma (PDAC) is highly resistant to conventional chemotherapeutic interventions, resulting in exceptionally low survival rates. The limited efficacy can in part be attributed to dose limitations and treatment cessation urged by toxicity of currently used chemotherapy. The advent of targeted delivery strategies has kindled hope for circumventing off-target toxicity. We have previously reported a PDAC-specific mesoporous silica nanoparticle (MSN) containing a protease linker responsive to ADAM9, a PDAC-enriched extracellularly deposited protease. Upon loading with paclitaxel these ADAM9-MSNs reduced side effects both in vitro and in vivo, however, disappointing antitumor efficacy was observed in vivo. Here, we propose that an efficient uptake of MSNs by tumor cells might underlie the lack of antitumor efficacy of MSNs functionalized with linker responsive to extracellular proteases. Harnessing this premise to improve antitumor efficacy, we performed an in silico analysis to identify PDAC-enriched intracellular proteases. We report the identification of BACE2, CAPN2 and DPP3 as PDAC enriched intracellular proteases, and report the synthesis of BACE2-, CAPN2- and DPP3-responsive MSNs. Extensive preclinical assessments revealed that paclitaxel-loaded CAPN2- and DPP3-MSNs exhibit high PDAC specificity in vitro as opposed to free paclitaxel. The administration of paclitaxel-loaded CAPN2- and DPP3-MSNs in vivo confirmed the reduction of leukopenia and induced no organ damage. Promisingly, in two mouse models CAPN2-MSNs reduced tumor growth at least as efficiently as free paclitaxel. Taken together, our results pose CAPN2-MSNs as a promising nanocarrier for the targeted delivery of chemotherapeutics in PDAC.
Collapse
Affiliation(s)
- Etienne J Slapak
- Amsterdam UMC Location University of Amsterdam, Center of Experimental and Molecular Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| | - Mouad El Mandili
- Amsterdam UMC Location University of Amsterdam, Center of Experimental and Molecular Medicine, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Marieke S Ten Brink
- Amsterdam UMC Location University of Amsterdam, Center of Experimental and Molecular Medicine, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Alexander Kros
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands.
| | - Maarten F Bijlsma
- Amsterdam UMC Location University of Amsterdam, Center of Experimental and Molecular Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| | - C Arnold Spek
- Amsterdam UMC Location University of Amsterdam, Center of Experimental and Molecular Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| |
Collapse
|
24
|
Mietzner R, Barbey C, Lehr H, Ziegler CE, Peterhoff D, Wagner R, Goepferich A, Breunig M. Prolonged delivery of HIV-1 vaccine nanoparticles from hydrogels. Int J Pharm 2024; 657:124131. [PMID: 38643811 DOI: 10.1016/j.ijpharm.2024.124131] [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: 02/13/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 04/23/2024]
Abstract
Immunization is a straightforward concept but remains for some pathogens like HIV-1 a challenge. Thus, new approaches towards increasing the efficacy of vaccines are required to turn the tide. There is increasing evidence that antigen exposure over several days to weeks induces a much stronger and more sustained immune response compared to traditional bolus injection, which usually leads to antigen elimination from the body within a couple of days. Therefore, we developed a poly(ethylene) glycol (PEG) hydrogel platform to investigate the principal feasibility of a sustained release of antigens to mimic natural infection kinetics. Eight-and four-armed PEG macromonomers of different MWs (10, 20, and 40 kDa) were end-group functionalized to allow for hydrogel formation via covalent cross-linking. An HIV-1 envelope (Env) antigen in its trimeric (Envtri) or monomeric (Envmono) form was applied. The soluble Env antigen was compared to a formulation of Env attached to silica nanoparticles (Env-SiNPs). The latter are known to have a higher immunogenicity compared to their soluble counterparts. Hydrogels were tunable regarding the rheological behavior allowing for different degradation times and release timeframes of Env-SiNPs over two to up to 50 days. Affinity measurements of the VCR01 antibody which specifically recognizes the CD4 binding site of Env, revealed that neither the integrity nor the functionality of Envmono-SiNPs (Kd = 2.1 ± 0.9 nM) and Envtri-SiNPs (Kd = 1.5 ± 1.3 nM), respectively, were impaired after release from the hydrogel (Kd before release: 2.1 ± 0.1 and 7.8 ± 5.3 nM, respectively). Finally, soluble Env and Env-SiNPs which are two physico-chemically distinct compounds, were co-delivered and shown to be sequentially released from one hydrogel which could be beneficial in terms of heterologous immunization or single dose vaccination. In summary, this study presents a tunable, versatile applicable, and effective delivery platform that could improve vaccination effectiveness also for other infectious diseases than HIV-1.
Collapse
Affiliation(s)
- Raphael Mietzner
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany
| | - Clara Barbey
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany
| | - Heike Lehr
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany
| | - Christian E Ziegler
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany
| | - David Peterhoff
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany.
| |
Collapse
|
25
|
Ghorbani R, Gharbavi M, Keshavarz B, Madanchi H, Johari B. Targeting c-Myc with decoy oligodeoxynucleotide-loaded polycationic nanoparticles inhibits cell growth and induces apoptosis in cancer stem-like cells (NTERA-2). Mol Biol Rep 2024; 51:623. [PMID: 38710891 DOI: 10.1007/s11033-024-09559-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 04/16/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND An increase in cancer stem cell (CSC) populations and their resistance to common treatments could be a result of c-Myc dysregulations in certain cancer cells. In the current study, we investigated anticancer effects of c-Myc decoy ODNs loaded-poly (methacrylic acid-co-diallyl dimethyl ammonium chloride) (PMA-DDA)-coated silica nanoparticles as carriers on cancer-like stem cells (NTERA-2). METHODS AND RESULTS The physicochemical characteristics of the synthesized nanocomposites (SiO2@PMA-DDA-DEC) were analyzed using FT-IR, DLS, and SEM techniques. UV-Vis spectrophotometer was applied to analyze the release pattern of decoy ODNs from the nanocomposite. Furthermore, uptake, cell viability, apoptosis, and cell cycle assays were used to investigate the anticancer effects of nanocomposites loaded with c-Myc decoy ODNs on NTERA-2 cancer cells. The results of physicochemical analytics demonstrated that SiO2@PMA-DDA-DEC nanocomposites were successfully synthesized. The prepared nanocomposites were taken up by NTERA-2 cells with high efficiency, and could effectively inhibit cell growth and increase apoptosis rate in the treated cells compared to the control group. Moreover, SiO2@PMA-DDA nanocomposites loaded with c-Myc decoy ODNs induced cell cycle arrest at the G0/G1 phase in the treated cells. CONCLUSIONS The conclusion drawn from this study is that c-Myc decoy ODN-loaded SiO2@PMA-DDA nanocomposites can effectively inhibit cell growth and induce apoptosis in NTERA-2 cancer cells. Moreover, given that a metal core is incorporated into this synthetic nanocomposite, it could potentially be used in conjunction with irradiation as part of a decoy-radiotherapy combinational therapy in future investigations.
Collapse
Affiliation(s)
- Roghayeh Ghorbani
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahmoud Gharbavi
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Pain Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Benyamin Keshavarz
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamid Madanchi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Biotechnology, School of Medicine, Semnan University of Medical Sciences, Semnan, 35131-38111, Iran.
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 13198, Iran.
| | - Behrooz Johari
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
26
|
Tang H, Wang Z, Hao H, Luo W, Yang J, Li M, Yang M, Chen Z, Yan R, Li H, Hu F, Liang H, Liu Q, Lv L, Zhang J, Su W, Chen R, Chen K, Chang YN, Wang M, Zheng L, Feng X, Li J, Xing G. Boron-Containing Mesoporous Silica Nanoparticles with Effective Delivery and Targeting of Liver Cancer Cells for Boron Neutron Capture Therapy. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38686647 DOI: 10.1021/acsami.4c02897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Nanocarriers have been researched comprehensively for the development of novel boron-containing agents in boron neutron capture therapy (BNCT). We designed and synthesized a multifunctional mesoporous silica nanoparticle (MSN)-based boron-containing agent. The latter was coated with a lipid bilayer (LB) and decorated with SP94 peptide (SFSIIHTPILPL) on the surface as SP94-LB@BA-MSN. The latter incorporated boric acid (BA) into hydrophobic mesopores, coated with an LB, and modified with SP94 peptide on the LB. SP94-LB@BA-MSN enhanced nano interface tumor-targeting ability but also prevented the premature release of drugs, which is crucial for BNCT because adequate boron content in tumor sites is required. SP94-LB@BA-MSN showed excellent efficacy in the BNCT treatment of HepG-2 cells. In animal studies with tumor-bearing mice, SP94-LB@BA-MSN exhibited a satisfactory accumulation at the tumor site. The boron content reached 40.18 ± 5.41 ppm in the tumor site 4 h after injection, which was 8.12 and 15.51 times higher than those in mice treated with boronated phenylalanine and those treated with BA. For boron, the tumor-to-normal tissue ratio was 4.41 ± 1.13 and the tumor-to-blood ratio was 5.92 ± 0.45. These results indicated that nanoparticles delivered boron to the tumor site effectively while minimizing accumulation in normal tissues. In conclusion, this composite (SP94-LB@BA-MSN) shows great promise as a boron-containing delivery agent for the treatment of hepatocellular carcinoma using BNCT. These findings highlight the potential of MSNs in the field of BNCT.
Collapse
Affiliation(s)
- Hongyu Tang
- School of Pharmacy, China Medical University, Shenyang 110122, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Zhijie Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Haoyang Hao
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Weixian Luo
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Jingru Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Mengyao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Mingxin Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Ziteng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Ruyu Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Hao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Fan Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Haojun Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Qiuyang Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Linwen Lv
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Junhui Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Wenxi Su
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Ranran Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Kui Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Ya-Nan Chang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Meng Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Lingna Zheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Xuesong Feng
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Juan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Gengmei Xing
- CAS Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| |
Collapse
|
27
|
Shahbaz S, Esmaeili M, Fathian Nasab MH, Imani Z, Bafkary R, Amini M, Atyabi F, Dinarvand R. PEGylated mesoporous silica core-shell redox-responsive nanoparticles for delivering paclitaxel to breast cancer cells. Int J Pharm 2024; 655:124024. [PMID: 38537920 DOI: 10.1016/j.ijpharm.2024.124024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024]
Abstract
Controlling the drug release and restricting its presence in healthy organs is extremely valuable. In this study, mesoporous silica nanoparticles (MSN) as the core, loaded with paclitaxel (PTX), were coated with a non-porous silica shell functionalized with disulfide bonds. The nanoparticles were further coated with polyethylene glycol (PEG) via disulfide linkages. We analyzed the physicochemical properties of nanoparticles, including hydrodynamic size via Dynamic Light Scattering (DLS), zeta potential, X-ray Diffraction (XRD) patterns, Fourier-Transform Infrared (FTIR) spectra, and imaging through Transmission Electron Microscopy (TEM) and Scanning Electron Microscopy (SEM). The drug release profile in two distinct glutathione (GSH) concentrations of 2 µM and 10 µM was measured. The cellular uptake of nanoparticles by MCF-7 cell line was determined using Confocal Laser Scanning Microscopy (CLSM) images and flow cytometry. Furthermore, the cell viability and the capability of nanoparticles to induce apoptosis in MCF-7 cell line were studied using the MTT assay and flow cytometry, respectively. Our investigations revealed that the release of PTX from the drug delivery system was redox-responsive. Also, results indicated an elevated level of cellular uptake and efficient induction of apoptosis, underscoring the promising potential of this redox-responsive drug delivery system for breast cancer therapy.
Collapse
Affiliation(s)
- Saeed Shahbaz
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahta Esmaeili
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Zhila Imani
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Bafkary
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Leicester School of Pharmacy, Leicester Institute for Pharmaceutical Innovation, De Montfort University, Leicester, UK.
| |
Collapse
|
28
|
Kovtareva S, Kusepova L, Tazhkenova G, Mashan T, Bazarbaeva K, Kopishev E. Surface Modification of Mesoporous Silica Nanoparticles for Application in Targeted Delivery Systems of Antitumour Drugs. Polymers (Basel) 2024; 16:1105. [PMID: 38675024 PMCID: PMC11054758 DOI: 10.3390/polym16081105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The problem of tumour therapy has attracted the attention of many researchers for many decades. One of the promising strategies for the development of new dosage forms to improve oncology treatment efficacy and minimise side effects is the development of nanoparticle-based targeted transport systems for anticancer drugs. Among inorganic nanoparticles, mesoporous silica deserves special attention due to its outstanding surface properties and drug-loading capability. This review analyses the various factors affecting the cytotoxicity, cellular uptake, and biocompatibility of mesoporous silica nanoparticles (MSNs), constituting a key aspect in the development of safe and effective drug delivery systems. Special attention is paid to technological approaches to chemically modifying MSNs to alter their surface properties. The stimuli that regulate drug release from nanoparticles are also discussed, contributing to the effective control of the delivery process in the body. The findings emphasise the importance of modifying MSNs with different surface functional groups, bio-recognisable molecules, and polymers for their potential use in anticancer drug delivery systems.
Collapse
Affiliation(s)
| | | | | | | | | | - Eldar Kopishev
- Department of Chemistry, Faculty of Natural Sciences, L.N. Gumilyov Eurasian National University, Astana 010000, Kazakhstan; (S.K.); (L.K.); (G.T.); (T.M.); (K.B.)
| |
Collapse
|
29
|
Yang H, Zhou JN, Zhang XM, Ling DD, Sun YB, Li CY, Zhou QQ, Shi GN, Wang SH, Lin XS, Fan T, Wang HY, Zeng Q, Jia YL, Xi JF, Jin YG, Pei XT, Yue W. Nanoengineered Red Blood Cells Loaded with TMPRSS2 and Cathepsin L Inhibitors Block SARS-CoV-2 Pseudovirus Entry into Lung ACE2 + Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310306. [PMID: 38194699 DOI: 10.1002/adma.202310306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/27/2023] [Indexed: 01/11/2024]
Abstract
The enzymatic activities of Furin, Transmembrane serine proteinase 2 (TMPRSS2), Cathepsin L (CTSL), and Angiotensin-converting enzyme 2 (ACE2) receptor binding are necessary for the entry of coronaviruses into host cells. Precise inhibition of these key proteases in ACE2+ lung cells during a viral infection cycle shall prevent viral Spike (S) protein activation and its fusion with a host cell membrane, consequently averting virus entry to the cells. In this study, dual-drug-combined (TMPRSS2 inhibitor Camostat and CTSL inhibitor E-64d) nanocarriers (NCs) are constructed conjugated with an anti-human ACE2 (hACE2) antibody and employ Red Blood Cell (RBC)-hitchhiking, termed "Nanoengineered RBCs," for targeting lung cells. The significant therapeutic efficacy of the dual-drug-loaded nanoengineered RBCs in pseudovirus-infected K18-hACE2 transgenic mice is reported. Notably, the modular nanoengineered RBCs (anti-receptor antibody+NCs+RBCs) precisely target key proteases of host cells in the lungs to block the entry of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), regardless of virus variations. These findings are anticipated to benefit the development of a series of novel and safe host-cell-protecting antiviral therapies.
Collapse
Affiliation(s)
- Hui Yang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jun-Nian Zhou
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xue-Mei Zhang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Dan-Dan Ling
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ying-Bao Sun
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chen-Yan Li
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Qian-Qian Zhou
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Gao-Na Shi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Si-Han Wang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xiao-Song Lin
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Tao Fan
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hai-Yang Wang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Quan Zeng
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ya-Li Jia
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jia-Fei Xi
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yi-Guang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xue-Tao Pei
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Wen Yue
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| |
Collapse
|
30
|
Yu J, Dan N, Eslami SM, Lu X. State of the Art of Silica Nanoparticles: An Overview on Biodistribution and Preclinical Toxicity Studies. AAPS J 2024; 26:35. [PMID: 38514482 DOI: 10.1208/s12248-024-00906-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
Over the past few years, nanoparticles have drawn particular attention in designing and developing drug delivery systems due to their distinctive advantages like improved pharmacokinetics, reduced toxicity, and specificity. Along with other successful nanosystems, silica nanoparticles (SNPs) have shown promising effects for therapeutic and diagnostic purposes. These nanoparticles are of great significance owing to their modifiable surface with various ligands, tunable particle size, and large surface area. The rate and extent of degradation and clearance of SNPs depend on factors such as size, shape, porosity, and surface modification, which directly lead to varying toxic mechanisms. Despite SNPs' enormous potential for clinical and pharmaceutical applications, safety concerns have hindered their translation into the clinic. This review discusses the biodistribution, toxicity, and clearance of SNPs and the formulation-related factors that ultimately influence clinical efficacy and safety for treatment. A holistic view of SNP safety will be beneficial for developing an enabling SNP-based drug product.
Collapse
Affiliation(s)
- Joshua Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Nirnoy Dan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Seyyed Majid Eslami
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Xiuling Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA.
| |
Collapse
|
31
|
Chen SY, Jian JY, Lin HM. Functionalization of rice husk-derived mesoporous silica nanoparticles for targeted and imaging in cancer drug delivery. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:2120-2129. [PMID: 38009620 DOI: 10.1002/jsfa.13165] [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: 05/23/2023] [Revised: 09/03/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Rice, a pivotal global food staple, annually accumulates vast amounts of rice husks, resulting in substantial environmental impact. Exploiting the high silica content in rice husk, our research aimed to recycle this agricultural byproduct to synthesize mesoporous silica nanoparticles (rMSNs). These nanoparticles were further modified to evaluate their potential as effective carriers for cancer drug delivery. RESULTS rMSNs showed high biocompatibility, large surface area and porous structure as MSNs, making them excellent drug carriers. Further modifications were applied to rMSNs, such as the incorporation of the lanthanides europium and gadolinium into rMSNs, making them fluorescent and magnetic for detection and tracking using confocal fluorescence microscopy and magnetic resonance imaging. Additionally, folic acid and aptamer AS1411 were conjugated with rMSNs to enhance the targeting of cancer cells. HeLa cells exhibited higher uptake of camptothecin (CPT)-loaded rMSNs compared to normal fibroblast cells (L929). The linkage of disulfide bonds to rMSNs also allowed CPT to be carried by rMSNs and released intracellularly in the presence of the abundant reducing agent glutathione. The validation of rMSNs in vitro and in vivo proved their practical feasibility. CONCLUSION Our findings indicate that low-cost rMSNs, derived from recycled agricultural waste, can replace highly valuable MSNs. Functionalized rMSNs exhibit promising capabilities in transporting clinical drugs to specific aberrant tissues and offering dual-targeting and dual-imaging functionalities for enhanced cancer therapy. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Shiow-Yi Chen
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City, Taiwan
| | - Jhih-Yun Jian
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City, Taiwan
| | - Hsiu-Mei Lin
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City, Taiwan
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung City, Taiwan
- Center of Excellence for Ocean Engineering, National Taiwan Ocean University, Keelung City, Taiwan
| |
Collapse
|
32
|
Kim K, Park MH. Advancing Cancer Treatment: Enhanced Combination Therapy through Functionalized Porous Nanoparticles. Biomedicines 2024; 12:326. [PMID: 38397928 PMCID: PMC10887220 DOI: 10.3390/biomedicines12020326] [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: 01/10/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Cancer remains a major global health challenge, necessitating the development of innovative treatment strategies. This review focuses on the functionalization of porous nanoparticles for combination therapy, a promising approach to enhance cancer treatment efficacy while mitigating the limitations associated with conventional methods. Combination therapy, integrating multiple treatment modalities such as chemotherapy, phototherapy, immunotherapy, and others, has emerged as an effective strategy to address the shortcomings of individual treatments. The unique properties of mesoporous silica nanoparticles (MSN) and other porous materials, like nanoparticles coated with mesoporous silica (NP@MS), metal-organic frameworks (MOF), mesoporous platinum nanoparticles (mesoPt), and carbon dots (CDs), are being explored for drug solubility, bioavailability, targeted delivery, and controlled drug release. Recent advancements in the functionalization of mesoporous nanoparticles with ligands, biomaterials, and polymers are reviewed here, highlighting their role in enhancing the efficacy of combination therapy. Various research has demonstrated the effectiveness of these nanoparticles in co-delivering drugs and photosensitizers, achieving targeted delivery, and responding to multiple stimuli for controlled drug release. This review introduces the synthesis and functionalization methods of these porous nanoparticles, along with their applications in combination therapy.
Collapse
Affiliation(s)
- Kibeom Kim
- Convergence Research Center, Nanobiomaterials Institute, Sahmyook University, Seoul 01795, Republic of Korea;
| | - Myoung-Hwan Park
- Convergence Research Center, Nanobiomaterials Institute, Sahmyook University, Seoul 01795, Republic of Korea;
- Department of Chemistry and Life Science, Sahmyook University, Seoul 01795, Republic of Korea
- Department of Convergence Science, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
33
|
Song SH, Bae M, Oh JK. Durable Surface Modification of Low-Density Polyethylene/Nano-Silica Composite Films with Bacterial Antifouling and Liquid-Repelling Properties for Food Hygiene and Safety. Polymers (Basel) 2024; 16:292. [PMID: 38276700 PMCID: PMC10819097 DOI: 10.3390/polym16020292] [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: 11/18/2023] [Revised: 01/14/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
The growing prevalence of antimicrobial resistance in bacterial strains has increased the demand for preventing biological deterioration on the surfaces of films used in applications involving food contact materials (FCMs). Herein, we prepared superhydrophobic film surfaces using a casting process that involved the combination of low-density polyethylene (LDPE) with solutions containing surface energy-reducing silica (SRS). The bacterial antifouling properties of the modified film surfaces were evaluated using Escherichia coli O157:H7 and Staphylococcus epidermidis via the dip-inoculation technique. The reduction in bacterial populations on the LDPE film embedded with SRS was confirmed to be more than 2 log-units, which equates to over 99%, when compared to the bare LDPE film. Additionally, the modified film demonstrated liquid-repelling properties against food-related contaminants, such as blood, beverages, and sauces. Moreover, the modified film demonstrated enhanced durability and robustness compared to one of the prevalent industry methods, dip-coating. We anticipate that the developed LDPE/nano-silica composite film represents a promising advancement in the multidisciplinary aspects of food hygiene and safety within the food industry, particularly concerning FCMs.
Collapse
Affiliation(s)
- Sang Ha Song
- Department of Polymer Science and Engineering, Dankook University, 152 Jukjeon-ro, Suji-gu, Yongin-si 16890, Republic of Korea;
| | - Michael Bae
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX 77845, USA
| | - Jun Kyun Oh
- Department of Polymer Science and Engineering, Dankook University, 152 Jukjeon-ro, Suji-gu, Yongin-si 16890, Republic of Korea;
| |
Collapse
|
34
|
Hwangbo H, Chae S, Kim W, Jo S, Kim GH. Tumor-on-a-chip models combined with mini-tissues or organoids for engineering tumor tissues. Theranostics 2024; 14:33-55. [PMID: 38164155 PMCID: PMC10750204 DOI: 10.7150/thno.90093] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/15/2023] [Indexed: 01/03/2024] Open
Abstract
The integration of tumor-on-a-chip technology with mini-tissues or organoids has emerged as a powerful approach in cancer research and drug development. This review provides an extensive examination of the diverse biofabrication methods employed to create mini-tissues, including 3D bioprinting, spheroids, microfluidic systems, and self-assembly techniques using cell-laden hydrogels. Furthermore, it explores various approaches for fabricating organ-on-a-chip platforms. This paper highlights the synergistic potential of combining these technologies to create tumor-on-a-chip models that mimic the complex tumor microenvironment and offer unique insights into cancer biology and therapeutic responses.
Collapse
Affiliation(s)
| | | | | | | | - Geun Hyung Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM) Suwon 16419, Republic of Korea
| |
Collapse
|
35
|
Mainuddin, Kumar A, Sharma A, Sharma N. Anti-cancer Drug Targeting using Stimuli Sensitive Mesoporous Silica Nanoparticle in Colorectal Cancer. Curr Pharm Des 2024; 30:3071-3073. [PMID: 39234908 DOI: 10.2174/0113816128321020240903100601] [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: 07/26/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Affiliation(s)
- Mainuddin
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Sector-125, Noida, 201313, Uttar Pradesh, India
| | - Anoop Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, 250005, India
| | - Anjana Sharma
- Department of Pharmaceutical Technology, Lloyd Institute of Management and Technology, Plot No. 11, Knowledge Park-II, Greater Noida, 201306, India
| | - Nitin Sharma
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Sector-125, Noida, 201313, Uttar Pradesh, India
| |
Collapse
|
36
|
Heidari R, Assadollahi V, Khosravian P, Mirzaei SA, Elahian F. Engineered mesoporous silica nanoparticles, new insight nanoplatforms into effective cancer gene therapy. Int J Biol Macromol 2023; 253:127060. [PMID: 37774811 DOI: 10.1016/j.ijbiomac.2023.127060] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/01/2023]
Abstract
The use of nucleic acid to control the expression of genes relevant to tumor progression is a key therapeutic approach in cancer research. Therapeutics based on nucleic acid provide novel concepts for untreatable targets. Nucleic acids as molecular medications must enter the target cell to be effective and obstacles in the systemic delivery of DNA or RNA limit their use in a clinical setting. The creation of nucleic acid delivery systems based on nanoparticles in order to circumvent biological constraints is advancing quickly. The ease of synthesis and surface modification, biocompatibility, biodegradability, cost-effectiveness and high loading capability of nucleic acids have prompted the use of mesoporous silica nanoparticles (MSNs) in gene therapy. The unique surface features of MSNs facilitate their design and decoration for high loading of nucleic acids, immune system evasion, cancer cell targeting, controlled cargo release, and endosomal escape. Reports have demonstrated successful therapeutic outcomes with the administration of a variety of engineered MSNs capable of delivering genes to tumor sites in laboratory animals. This comprehensive review of studies about siRNA, miRNA, shRNA, lncRNA and CRISPR/Cas9 delivery by MSNs reveals engineered MSNs as a safe and efficient system for gene transfer to cancer cells and cancer mouse models.
Collapse
Affiliation(s)
- Razieh Heidari
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Vahideh Assadollahi
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Pegah Khosravian
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Abbas Mirzaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran; Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Elahian
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; Human Stem Cells and Neuronal Differentiation Core, Baylor College of Medicine, Houston, USA.
| |
Collapse
|
37
|
Janjua TI, Cao Y, Kleitz F, Linden M, Yu C, Popat A. Silica nanoparticles: A review of their safety and current strategies to overcome biological barriers. Adv Drug Deliv Rev 2023; 203:115115. [PMID: 37844843 DOI: 10.1016/j.addr.2023.115115] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023]
Abstract
Silica nanoparticles (SNP) have gained tremendous attention in the recent decades. They have been used in many different biomedical fields including diagnosis, biosensing and drug delivery. Medical uses of SNP for anti-cancer, anti-microbial and theranostic applications are especially prominent due to their exceptional performance to deliver many different small molecules and recently biologics (mRNA, siRNA, antigens, antibodies, proteins, and peptides) at targeted sites. The physical and chemical properties of SNP such as large specific surface area, tuneable particle size and porosity, excellent biodegradability and biocompatibility make them an ideal drug delivery and diagnostic platform. Based on the available data and the pre-clinical performance of SNP, recent interest has driven these innovative materials towards clinical application with many of the formulations already in Phase I and Phase II trials. Herein, the progress of SNP in biomedical field is reviewed, and their safety aspects are analysed. Importantly, we critically evaluate the key structural characteristics of SNP to overcome different biological barriers including the blood-brain barrier (BBB), skin, tumour barrier and mucosal barrier. Future directions, potential pathways, and target areas towards rapid clinical translation of SNP are also recommended.
Collapse
Affiliation(s)
- Taskeen Iqbal Janjua
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| | - Yuxue Cao
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Freddy Kleitz
- Department of Functional Materials and Catalysis, Faculty of Chemistry, University of Vienna, Währinger Straße 42, 1090 Vienna, Austria
| | - Mika Linden
- Institute of Inorganic Chemistry II, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Queensland, QLD 4072, Australia.
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia; Department of Functional Materials and Catalysis, Faculty of Chemistry, University of Vienna, Währinger Straße 42, 1090 Vienna, Austria.
| |
Collapse
|
38
|
Kaymaz SV, Nobar HM, Sarıgül H, Soylukan C, Akyüz L, Yüce M. Nanomaterial surface modification toolkit: Principles, components, recipes, and applications. Adv Colloid Interface Sci 2023; 322:103035. [PMID: 37931382 DOI: 10.1016/j.cis.2023.103035] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/11/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023]
Abstract
Surface-functionalized nanostructures are at the forefront of biotechnology, providing new opportunities for biosensors, drug delivery, therapy, and bioimaging applications. The modification of nanostructures significantly impacts the performance and success of various applications by enabling selective and precise targeting. This review elucidates widely practiced surface modification strategies, including click chemistry, cross-coupling, silanization, aldehyde linkers, active ester chemistry, maleimide chemistry, epoxy linkers, and other protein and DNA-based methodologies. We also delve into the application-focused landscape of the nano-bio interface, emphasizing four key domains: therapeutics, biosensing, environmental monitoring, and point-of-care technologies, by highlighting prominent studies. The insights presented herein pave the way for further innovations at the intersection of nanotechnology and biotechnology, providing a useful handbook for beginners and professionals. The review draws on various sources, including the latest research articles (2018-2023), to provide a comprehensive overview of the field.
Collapse
Affiliation(s)
- Sümeyra Vural Kaymaz
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey; SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| | | | - Hasan Sarıgül
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| | - Caner Soylukan
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| | - Lalehan Akyüz
- Department of Molecular Biology and Genetics, Aksaray University, 68100 Aksaray, Turkey
| | - Meral Yüce
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey.
| |
Collapse
|
39
|
Sivamaruthi BS, Kapoor DU, Kukkar RR, Gaur M, Elossaily GM, Prajapati BG, Chaiyasut C. Mesoporous Silica Nanoparticles: Types, Synthesis, Role in the Treatment of Alzheimer's Disease, and Other Applications. Pharmaceutics 2023; 15:2666. [PMID: 38140007 PMCID: PMC10747102 DOI: 10.3390/pharmaceutics15122666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/25/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Globally, many individuals struggle with Alzheimer's disease (AD), an unrelenting and incapacitating neurodegenerative condition. Despite notable research endeavors, effective remedies for AD remain constrained, prompting the exploration of innovative therapeutic avenues. Within this context, silica-based nanoplatforms have emerged with pronounced potential due to their unique attributes like expansive surface area, customizable pore dimensions, and compatibility with living systems. These nanoplatforms hold promise as prospective interventions for AD. This assessment provides a comprehensive overview encompassing various forms of mesoporous silica nanoparticles (MSNs), techniques for formulation, and their applications in biomedicine. A significant feature lies in their ability to precisely guide and control the transport of therapeutic agents to the brain, facilitated by the adaptability of these nanoplatforms as drug carriers. Their utility as tools for early detection and monitoring of AD is investigated. Challenges and prospects associated with harnessing MSNs are studied, underscoring the imperative of stringent safety evaluations and optimization of how they interact with the body. Additionally, the incorporation of multifunctional attributes like imaging and targeting components is emphasized to enhance their efficacy within the intricate milieu of AD. As the battle against the profound repercussions of AD persists, MSNs emerge as a promising avenue with the potential to propel the development of viable therapeutic interventions.
Collapse
Affiliation(s)
- Bhagavathi Sundaram Sivamaruthi
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand;
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Devesh U. Kapoor
- Department of Pharmacy, Dr. Dayaram Patel Pharmacy College, Bardoli 394601, Gujarat, India;
| | - Rajiv R. Kukkar
- School of Pharmacy, Raffles University, Neemrana 301705, Rajasthan, India
| | - Mansi Gaur
- Rajasthan Pharmacy College, Rajasthan University of Health Sciences, Jaipur 302033, Rajasthan, India
| | - Gehan M. Elossaily
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia;
| | - Bhupendra G. Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Mehsana 384012, Gujarat, India
| | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
40
|
Ramezanian S, Moghaddas J, Roghani-Mamaqani H, Rezamand A. Dual pH- and temperature-responsive poly(dimethylaminoethyl methacrylate)-coated mesoporous silica nanoparticles as a smart drug delivery system. Sci Rep 2023; 13:20194. [PMID: 37980442 PMCID: PMC10657431 DOI: 10.1038/s41598-023-47026-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/08/2023] [Indexed: 11/20/2023] Open
Abstract
A robust drug delivery system was created by grafting poly(dimethylaminoethyl methacrylate) (PDMAEMA) onto silica nanoparticles with two different lengths using an in situ atom transfer radical polymerization, resulting in the formation of a pH- and temperature-sensitive shell. The high molecular weight PDMAEMA demonstrated effective controlled drug release, and prevented drug release in healthy cells. Drug release occurred through polymer shell protonation at pH 5. The critical temperature of 41 °C facilitated rapid solvation of the shell polymers in the blood, preventing tissue accumulation and reducing toxicity compared to systems with lower critical solution temperatures. Field-emission scanning electron microscopy analysis and nitrogen adsorption/desorption analysis showed that the nanoparticles have a fine network, mesoporous structure, and a mean size of around 17 nm that show their excellent capacity for loading drugs. Fourier-transform infrared spectroscopy showed that all the modification steps and polymerization were successfully implemented. Thermogravimetric analysis showed PDMAEMA chains with two different lengths grafted onto the nanoparticles. Transmission electron microscopy analysis also showed grafted polymer chains on the hybrid nanoparticles. The release profile of model cancer drugs (doxorubicin and methotrexate) varied with pH and temperature, with high molecular weight PDMAEMA shells effectively preventing drug release at neutral pH. In vitro analysis using the HeLa cell line showed minimal toxicity in blank samples and significant release profile in acidic environment.
Collapse
Affiliation(s)
- Sina Ramezanian
- Chemical Engineering Faculty, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran
- Transport Phenomena Research Center, Chemical Engineering Faculty, Sahand University of Technology, P.O. Box 51335/1996, Tabriz, Iran
| | - Jafarsadegh Moghaddas
- Chemical Engineering Faculty, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran.
- Transport Phenomena Research Center, Chemical Engineering Faculty, Sahand University of Technology, P.O. Box 51335/1996, Tabriz, Iran.
| | - Hossein Roghani-Mamaqani
- Faculty of Polymer Engineering, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran
- Institute of Polymeric Materials, Sahand University of Technology, P.O. Box 51335-1996, Tabriz, Iran
| | - Azim Rezamand
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pediatrics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Barguilla I, Candela-Noguera V, Oliver P, Annangi B, Díez P, Aznar E, Martínez-Máñez R, Marcos R, Hernández A, Marcos MD. Toxicological Profiling and Long-Term Effects of Bare, PEGylated- and Galacto-Oligosaccharide-Functionalized Mesoporous Silica Nanoparticles. Int J Mol Sci 2023; 24:16158. [PMID: 38003350 PMCID: PMC10671840 DOI: 10.3390/ijms242216158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Mesoporous silica nanoparticles (MSNs) are amongst the most used nanoparticles in biomedicine. However, the potentially toxic effects of MSNs have not yet been fully evaluated, being a controversial matter in research. In this study, bare MSNs, PEGylated MSNs (MSNs-PEG), and galacto-oligosaccharide-functionalized MSNs (MSNs-GAL) are synthesized and characterized to assess their genotoxicity and transforming ability on human lung epithelial BEAS-2B cells in short- (48 h) and long-term (8 weeks) exposure scenarios. Initial short-term treatments show a dose-dependent increase in genotoxicity for MSNs-PEG-treated cells but not oxidative DNA damage for MSNs, MSNs-PEG, or for MSNs-GAL. In addition, after 8 weeks of continuous exposure, neither induced genotoxic nor oxidative DNA is observed. Nevertheless, long-term treatment with MSNs-PEG and MSNs-GAL, but not bare MSNs, induces cell transformation features, as evidenced by the cell's enhanced ability to grow independently of anchorage, to migrate, and to invade. Further, the secretome from cells treated with MSNs and MSNs-GAL, but not MSNs-PEG, shows certain tumor-promoting abilities, increasing the number and size of HeLa cell colonies formed in the indirect soft-agar assay. These results show that MSNs, specifically the functionalized ones, provoke some measurable adverse effects linked to tumorigenesis. These effects are in the order of other nanomaterials, such as carbon nanotubes or cerium dioxide nanoparticles, but they are lower than those provoked by some approved drugs, such as doxorubicin or dexamethasone.
Collapse
Affiliation(s)
- Irene Barguilla
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain (R.M.)
| | - Vicente Candela-Noguera
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, 46022 Valencia, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
| | - Patrick Oliver
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain (R.M.)
| | - Balasubramanyam Annangi
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain (R.M.)
| | - Paula Díez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, 46022 Valencia, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Instituto de Investigación Sanitaria La Fe (IISLAFE), Universitat Politècnica de València, 46026 Valencia, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Elena Aznar
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, 46022 Valencia, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Instituto de Investigación Sanitaria La Fe (IISLAFE), Universitat Politècnica de València, 46026 Valencia, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, 46022 Valencia, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Instituto de Investigación Sanitaria La Fe (IISLAFE), Universitat Politècnica de València, 46026 Valencia, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ricard Marcos
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain (R.M.)
| | - Alba Hernández
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain (R.M.)
| | - María Dolores Marcos
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, 46022 Valencia, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Instituto de Investigación Sanitaria La Fe (IISLAFE), Universitat Politècnica de València, 46026 Valencia, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
42
|
García-Fernández A, Sancho M, Garrido E, Bisbal V, Sancenón F, Martínez-Máñez R, Orzáez M. Targeted Delivery of the Pan-Inflammasome Inhibitor MM01 as an Alternative Approach to Acute Lung Injury Therapy. Adv Healthc Mater 2023; 12:e2301577. [PMID: 37515468 DOI: 10.1002/adhm.202301577] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/14/2023] [Indexed: 07/30/2023]
Abstract
Acute lung injury (ALI) is a severe pulmonary disorder responsible for high percentage of mortality and morbidity in intensive care unit patients. Current treatments are ineffective, so the development of efficient and specific therapies is an unmet medical need. The activation of NLPR3 inflammasome during ALI produces the release of proinflammatory factors and pyroptosis, a proinflammatory form of cell death that contributes to lung damage spreading. Herein, it is demonstrated that modulating inflammasome activation through inhibition of ASC oligomerization by the recently described MM01 compound can be an alternative pharmacotherapy against ALI. Besides, the added efficacy of using a drug delivery nanosystem designed to target the inflamed lungs is determined. The MM01 drug is incorporated into mesoporous silica nanoparticles capped with a peptide (TNFR-MM01-MSNs) to target tumor necrosis factor receptor-1 (TNFR-1) to proinflammatory macrophages. The prepared nanoparticles can deliver the cargo in a controlled manner after the preferential uptake by proinflammatory macrophages and exhibit anti-inflammatory activity. Finally, the therapeutic effect of MM01 free or nanoparticulated to inhibit inflammatory response and lung injury is successfully demonstrated in lipopolysaccharide-mouse model of ALI. The results suggest the potential of pan-inflammasome inhibitors as candidates for ALI therapy and the use of nanoparticles for targeted lung delivery.
Collapse
Affiliation(s)
- Alba García-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camí de vera s/n, Valencia, 46022, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5, Madrid, 28029, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, Valencia, 46012, Spain
| | - Mónica Sancho
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, Valencia, 46012, Spain
- Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera 3, Valencia, 46012, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat de València, Burjassot, E-46100, Spain
| | - Eva Garrido
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camí de vera s/n, Valencia, 46022, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5, Madrid, 28029, Spain
| | - Viviana Bisbal
- Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera 3, Valencia, 46012, Spain
| | - Félix Sancenón
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camí de vera s/n, Valencia, 46022, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5, Madrid, 28029, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, Valencia, 46012, Spain
- Departamento de Química, Universitat Politècnica de València, Camino de Vera s/n, Valencia, 46022, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, Valencia, 46026, Spain
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camí de vera s/n, Valencia, 46022, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5, Madrid, 28029, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, Valencia, 46012, Spain
- Departamento de Química, Universitat Politècnica de València, Camino de Vera s/n, Valencia, 46022, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, Valencia, 46026, Spain
| | - Mar Orzáez
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, Valencia, 46012, Spain
- Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera 3, Valencia, 46012, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat de València, Burjassot, E-46100, Spain
| |
Collapse
|
43
|
Malekian F, Shamsian A, Kodam SP, Ullah M. Exosome engineering for efficient and targeted drug delivery: Current status and future perspective. J Physiol 2023; 601:4853-4872. [PMID: 35570717 DOI: 10.1113/jp282799] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2023] Open
Abstract
Exosomes are membrane-bound vesicles that are released by most cells. They carry nucleic acids, cytokines, growth factors, proteins, lipids, and metabolites. They are responsible for inter- and intracellular communications and their role in drug delivery is well defined. Exosomes have great potential for therapeutic applications, but the clinical use is restricted because of limitations in standardized procedures for isolation, purification, and drug delivery. Bioengineering of exosomes could be one approach to achieve standardization and reproducible isolation for clinical use. Exosomes are important transporters for targeted drug delivery because of their small size, stable structure, non-immunogenicity, and non-toxic nature, as well as their ability to carry a wide variety of compounds. These features of exosomes can be enhanced further by bioengineering. In this review, possible exosome bioengineering approaches, their biomedical applications, and targeted drug delivery are discussed.
Collapse
Affiliation(s)
- Farzaneh Malekian
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Alireza Shamsian
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Sai Priyanka Kodam
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Mujib Ullah
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
- Molecular Medicine Department of Medicine, Stanford University, Palo Alto, CA, USA
| |
Collapse
|
44
|
Rehan T, Tahir A, Sultan A, Alabbosh KF, Waseem S, Ul-Islam M, Khan KA, Ibrahim EH, Ullah MW, Shah N. Mitigation of Benzene-Induced Haematotoxicity in Sprague Dawley Rats through Plant-Extract-Loaded Silica Nanobeads. TOXICS 2023; 11:865. [PMID: 37888715 PMCID: PMC10610980 DOI: 10.3390/toxics11100865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023]
Abstract
Benzene, a potent carcinogen, is known to cause acute myeloid leukaemia. While chemotherapy is commonly used for cancer treatment, its side effects have prompted scientists to explore natural products that can mitigate the haematotoxic effects induced by chemicals. One area of interest is nano-theragnostics, which aims to enhance the therapeutic potential of natural products. This study aimed to enhance the effects of methanolic extracts from Ocimum basilicum, Rosemarinus officinalis, and Thymus vulgaris by loading them onto silica nanobeads (SNBs) for targeted delivery to mitigate the benzene-induced haematotoxic effects. The SNBs, 48 nm in diameter, were prepared using a chemical method and were then loaded with the plant extracts. The plant-extract-loaded SNBs were then coated with carboxymethyl cellulose (CMC). The modified SNBs were characterized using various techniques such as scanning electron microscopy (SEM), X-ray diffraction (XRD), UV-visible spectroscopy, and Fourier transform infrared (FTIR) spectroscopy. The developed plant-extract-loaded and CMC-modified SNBs were administered intravenously to benzene-exposed rats, and haematological and histopathological profiling was conducted. Rats exposed to benzene showed increased liver and spleen weight, which was mitigated by the plant-extract-loaded SNBs. The differential white blood cell (WBC) count was higher in rats with benzene-induced haematotoxicity, but this count decreased significantly in rats treated with plant-extract-loaded SNBs. Additionally, blast cells observed in benzene-exposed rats were not found in rats treated with plant-extract-loaded SNBs. The SNBs facilitated targeted drug delivery of the three selected medicinal herbs at low doses. These results suggest that SNBs have promising potential as targeted drug delivery agents to mitigate haematotoxic effects induced by benzene in rats.
Collapse
Affiliation(s)
- Touseef Rehan
- Department of Biochemistry, Women University Mardan, Mardan 23200, Pakistan
| | - Anum Tahir
- Department of Biochemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Aneesa Sultan
- Department of Biochemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | | | - Shahid Waseem
- Department of Biochemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Mazhar Ul-Islam
- Department of Chemical Engineering, College of Engineering, Dhofar University, Salalah 211, Oman
| | - Khalid Ali Khan
- Unit of Bee Research and Honey Production, Research Center for
Advanced Materials Science (RCAMS), Applied College, King Khalid University, Abha 61413, Saudi Arabia
| | - Essam H. Ibrahim
- Biology Department, Faculty of Science, Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 61413, Saudi Arabia
- Blood Products Quality Control and Research Department, National Organization for Research and Control of Biologicals, Cairo 12611, Egypt
| | - Muhammad Wajid Ullah
- Biofuels Institute, School of the Environmental and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Nasrullah Shah
- Department of Chemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| |
Collapse
|
45
|
Chehelgerdi M, Chehelgerdi M, Allela OQB, Pecho RDC, Jayasankar N, Rao DP, Thamaraikani T, Vasanthan M, Viktor P, Lakshmaiya N, Saadh MJ, Amajd A, Abo-Zaid MA, Castillo-Acobo RY, Ismail AH, Amin AH, Akhavan-Sigari R. Progressing nanotechnology to improve targeted cancer treatment: overcoming hurdles in its clinical implementation. Mol Cancer 2023; 22:169. [PMID: 37814270 PMCID: PMC10561438 DOI: 10.1186/s12943-023-01865-0] [Citation(s) in RCA: 269] [Impact Index Per Article: 134.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 09/21/2023] [Indexed: 10/11/2023] Open
Abstract
The use of nanotechnology has the potential to revolutionize the detection and treatment of cancer. Developments in protein engineering and materials science have led to the emergence of new nanoscale targeting techniques, which offer renewed hope for cancer patients. While several nanocarriers for medicinal purposes have been approved for human trials, only a few have been authorized for clinical use in targeting cancer cells. In this review, we analyze some of the authorized formulations and discuss the challenges of translating findings from the lab to the clinic. This study highlights the various nanocarriers and compounds that can be used for selective tumor targeting and the inherent difficulties in cancer therapy. Nanotechnology provides a promising platform for improving cancer detection and treatment in the future, but further research is needed to overcome the current limitations in clinical translation.
Collapse
Affiliation(s)
- Mohammad Chehelgerdi
- Novin Genome (NG) Institute, Research and Development Center for Biotechnology, Shahrekord, Chaharmahal and Bakhtiari, Iran.
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Chaharmahal and Bakhtiari, Iran.
| | - Matin Chehelgerdi
- Novin Genome (NG) Institute, Research and Development Center for Biotechnology, Shahrekord, Chaharmahal and Bakhtiari, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Chaharmahal and Bakhtiari, Iran
| | | | | | - Narayanan Jayasankar
- Department of Pharmacology, SRM Institute of Science and Technology, SRM College Of Pharmacy, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Devendra Pratap Rao
- Department of Chemistry, Coordination Chemistry Laboratory, Dayanand Anglo-Vedic (PG) College, Kanpur-208001, U.P, India
| | - Tamilanban Thamaraikani
- Department of Pharmacology, SRM Institute of Science and Technology, SRM College Of Pharmacy, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Manimaran Vasanthan
- Department of Pharmaceutics, SRM Institute of Science and Technology, SRM College Of Pharmacy, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Patrik Viktor
- Keleti Károly Faculty of Business and Management, Óbuda University, Tavaszmező U. 15-17, 1084, Budapest, Hungary
| | - Natrayan Lakshmaiya
- Department of Mechanical Engineering, Saveetha School of Engineering, SIMATS, Chennai, Tamil Nadu, India
| | - Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Ayesha Amajd
- Faculty of Organization and Management, Silesian University of Technology, 44-100, Gliwice, Poland
- Department of Mechanical Engineering, CEMMPRE, University of Coimbra, Polo II, 3030-788, Coimbra, Portugal
| | - Mabrouk A Abo-Zaid
- Department of Biology, College of Science, Jazan University, 82817, Jazan, Saudi Arabia
| | | | - Ahmed H Ismail
- Department of Biology, College of Science, Jazan University, 82817, Jazan, Saudi Arabia
| | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warsaw, Poland
| |
Collapse
|
46
|
Shchaslyvyi AY, Antonenko SV, Tesliuk MG, Telegeev GD. Current State of Human Gene Therapy: Approved Products and Vectors. Pharmaceuticals (Basel) 2023; 16:1416. [PMID: 37895887 PMCID: PMC10609992 DOI: 10.3390/ph16101416] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
In the realm of gene therapy, a pivotal moment arrived with Paul Berg's groundbreaking identification of the first recombinant DNA in 1972. This achievement set the stage for future breakthroughs. Conditions once considered undefeatable, like melanoma, pancreatic cancer, and a host of other ailments, are now being addressed at their root cause-the genetic level. Presently, the gene therapy landscape stands adorned with 22 approved in vivo and ex vivo products, including IMLYGIC, LUXTURNA, Zolgensma, Spinraza, Patisiran, and many more. In this comprehensive exploration, we delve into a rich assortment of 16 drugs, from siRNA, miRNA, and CRISPR/Cas9 to DNA aptamers and TRAIL/APO2L, as well as 46 carriers, from AAV, AdV, LNPs, and exosomes to naked mRNA, sonoporation, and magnetofection. The article also discusses the advantages and disadvantages of each product and vector type, as well as the current challenges faced in the practical use of gene therapy and its future potential.
Collapse
Affiliation(s)
- Aladdin Y. Shchaslyvyi
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 150, Zabolotnogo Str., 03143 Kyiv, Ukraine; (S.V.A.); (M.G.T.); (G.D.T.)
| | | | | | | |
Collapse
|
47
|
Rathnayake K, Patel U, Hunt EC, Singh N. Fabrication of a Dual-Targeted Liposome-Coated Mesoporous Silica Core-Shell Nanoassembly for Targeted Cancer Therapy. ACS OMEGA 2023; 8:34481-34498. [PMID: 37779923 PMCID: PMC10536893 DOI: 10.1021/acsomega.3c02901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023]
Abstract
Nanoparticles have been suggested as drug-delivery systems for chemotherapeutic drugs to allow for controlled drug release profiles and selectivity to target cancer cells. In addition, nanoparticles can be used for the in situ generation and amplification of reactive oxygen species (ROS), which have been shown to be a promising strategy for cancer treatment. Thus, a targeted nanoscale drug-delivery platform could be used to synergistically improve cancer treatment by the action of chemotherapeutic drugs and ROS generation. Herein, we propose a promising chemotherapy strategy where the drug-loaded nanoparticles generate high doses of ROS together with the loaded ROS-generating chemotherapeutic drugs, which can damage the mitochondria and activate cell death, potentiating the therapeutic outcome in cancer therapy. In the present study, we have developed a dual-targeted drug-delivery nanoassembly consisting of a mesoporous silica core loaded with the chemotherapeutic, ROS-generating drug, paclitaxel (Px), and coated with a liposome layer for controlled drug release. Two different lung cancer-targeting ligands, folic acid and peptide GE11, were used to target the overexpressed nonsmall lung cancer receptors to create the final nanoassembly (MSN@Px) L-GF. Upon endocytosis by the cancer cells, the liposome layer was degraded by the intracellular lipases, and the drug was rapidly released at a rate of 65% within the first 20 h. In vitro studies confirmed that this nanoassembly was 8-fold more effective in cancer therapy compared to the free drug Px.
Collapse
Affiliation(s)
- Kavini Rathnayake
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Unnati Patel
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Emily C. Hunt
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Nirupama Singh
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| |
Collapse
|
48
|
Sultana N, David AE. Improving Cancer Targeting: A Study on the Effect of Dual-Ligand Density on Targeting of Cells Having Differential Expression of Target Biomarkers. Int J Mol Sci 2023; 24:13048. [PMID: 37685852 PMCID: PMC10487485 DOI: 10.3390/ijms241713048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Silica nanoparticles with hyaluronic acid (HA) and folic acid (FA) were developed to study dual-ligand targeting of CD44 and folate receptors, respectively, in colon cancer. Characterization of particles with dynamic light scattering showed them to have hydrodynamic diameters of 147-271 nm with moderate polydispersity index (PDI) values. Surface modification of the particles was achieved by simultaneous reaction with HA and FA and results showed that ligand density on the surface increased with increasing concentrations in the reaction mixture. The nanoparticles showed minimal to no cytotoxicity with all formulations showing ≥ 90% cell viability at concentrations up to 100 µg/mL. Based on flow cytometry results, SW480 cell lines were positive for both receptors, the WI38 cell line was positive for CD44 receptor, and Caco2 was positive for the folate receptor. Cellular targeting studies demonstrated the potential of the targeted nanoparticles as promising candidates for delivery of therapeutic agents. The highest cellular targeting was achieved with particles synthesized using folate:surface amine (F:A) ratio of 9 for SW480 and Caco2 cells and at F:A = 0 for WI38 cells. The highest selectivity was achieved at F:A = 9 for both SW480:WI38 and SW480:Caco2 cells. Based on HA conjugation, the highest cellular targeting was achieved at H:A = 0.5-0.75 for SW480 cell, at H:A = 0.75 for WI38 cell and at H:A = 0.5 for Caco2 cells. The highest selectivity was achieved at H:A = 0 for both SW480:WI38 and SW480:Caco2 cells. These results demonstrated that the optimum ligand density on the nanoparticle for targeting is dependent on the levels of biomarker expression on the target cells. Ongoing studies will evaluate the therapeutic efficacy of these targeted nanoparticles using in vitro and in vivo cancer models.
Collapse
Affiliation(s)
| | - Allan E. David
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA;
| |
Collapse
|
49
|
Eckenberger E, Raczka T, Neuhuber W, Distel LVR, Klein S. Acriflavine-Functionalized Silica@Manganese Ferrite Nanostructures for Synergistic Radiation and Hypoxia Therapies. ACS APPLIED BIO MATERIALS 2023; 6:3089-3102. [PMID: 37433114 DOI: 10.1021/acsabm.2c01021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Mesoporous and nonmesoporous SiO2@MnFe2O4 nanostructures were loaded with the hypoxia-inducible factor-1 inhibitor acriflavine for combined radiation and hypoxia therapies. The X-ray irradiation of the drug-loaded nanostructures not only triggered the release of the acriflavine inside the cells but also initiated an energy transfer from the nanostructures to surface-adsorbed oxygen to generate singlet oxygen. While the drug-loaded mesoporous nanostructures showed an initial drug release before the irradiation, the drug was primarily released upon X-ray radiation in the case of the nonmesoporous nanostructures. However, the drug loading capacity was less efficient for the nonmesoporous nanostructures. Both drug-loaded nanostructures proved to be very efficient in irradiated MCF-7 multicellular tumor spheroids. The damage of these nanostructures toward the nontumorigenic MCF-10A multicellular spheroids was very limited because of the small number of nanostructures that entered the MCF-10A spheroids, while similar concentrations of acriflavine without nanostructures were toxic for the MCF-10A spheroids.
Collapse
Affiliation(s)
- Elisabeth Eckenberger
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr.3, D-91058 Erlangen, Germany
| | - Theodor Raczka
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr.3, D-91058 Erlangen, Germany
| | - Winfried Neuhuber
- Institute of Anatomy and Cell Biology, Chair of Anatomy I, Friedrich-Alexander University Erlangen-Nuremberg, Krankenhausstr. 9, D-91054 Erlangen, Germany
| | - Luitpold V R Distel
- Department of Radiation Oncology, Friedrich-Alexander University of Erlangen-Nuremberg, Universitätsstr. 27, D-91054 Erlangen, Germany
| | - Stefanie Klein
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr.3, D-91058 Erlangen, Germany
| |
Collapse
|
50
|
Garrido-Cano I, Adam-Artigues A, Lameirinhas A, Blandez JF, Candela-Noguera V, Lluch A, Bermejo B, Sancenón F, Cejalvo JM, Martínez-Máñez R, Eroles P. Delivery of miR-200c-3p Using Tumor-Targeted Mesoporous Silica Nanoparticles for Breast Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:38323-38334. [PMID: 37549382 PMCID: PMC10436244 DOI: 10.1021/acsami.3c07541] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/25/2023] [Indexed: 08/09/2023]
Abstract
Despite advances in breast cancer treatment, it remains the leading cause of cancer-related death in women worldwide. In this context, microRNAs have emerged as potential therapeutic targets but still present some limitations for in vivo applications. Particularly, miR-200c-3p is a well-known tumor suppressor microRNA that inhibits tumor progression and metastasis in breast cancer through downregulating ZEB1 and ZEB2. Based on the above, we describe the design and validation of a nanodevice using mesoporous silica nanoparticles for miR-200c-3p delivery for breast cancer treatment. We demonstrate the biocompatibility of the synthesized nanodevices as well as their ability to escape from endosomes/lysosomes and inhibit tumorigenesis, invasion, migration, and proliferation of tumor cells in vitro. Moreover, tumor targeting and effective delivery of miR-200c-3p from the nanoparticles in vivo are confirmed in an orthotopic breast cancer mouse model, and the therapeutic efficacy is also evidenced by a decrease in tumor size and lung metastasis, while showing no signs of toxicity. Overall, our results provide evidence that miR-200c-3p-loaded nanoparticles are a potential strategy for breast cancer therapy and a safe and effective system for tumor-targeted delivery of microRNAs.
Collapse
Affiliation(s)
- Iris Garrido-Cano
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
| | | | - Ana Lameirinhas
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
| | - Juan F. Blandez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València,
IIS La Fe, Valencia 46026, Spain
| | - Vicente Candela-Noguera
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
| | - Ana Lluch
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Universitat
de València, Valencia 46010, Spain
- Clinical
Oncology Department, Hospital Clínico
Universitario de Valencia, Valencia 46010, Spain
| | - Begoña Bermejo
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Clinical
Oncology Department, Hospital Clínico
Universitario de Valencia, Valencia 46010, Spain
| | - Felix Sancenón
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València,
IIS La Fe, Valencia 46026, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina. Universitat Politècnica de Valencia, Centro de Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Juan Miguel Cejalvo
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Clinical
Oncology Department, Hospital Clínico
Universitario de Valencia, Valencia 46010, Spain
| | - Ramón Martínez-Máñez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat Politècnica
de València, Universitat de València, Valencia 46010, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València,
IIS La Fe, Valencia 46026, Spain
- Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina. Universitat Politècnica de Valencia, Centro de Investigación Príncipe Felipe, Valencia 46012, Spain
| | - Pilar Eroles
- Biomedical
Research Institute INCLIVA, Valencia 46010, Spain
- Centro
de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid 28029, Spain
- Universitat
de València, Valencia 46010, Spain
| |
Collapse
|