1
|
Qasim Almajidi Y, Jawad AQ, Abdulameer Albadri A. Biocompatible PAMAM-PLGA-PCL Nanocarrier for Efficient Curcumin Delivery to Lung Cancer Cells: In Vitro Studies. Chem Biodivers 2024; 21:e202401106. [PMID: 39012926 DOI: 10.1002/cbdv.202401106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/18/2024]
Abstract
Lung cancer, as the leading cause of death among other types of cancer, has a high rate of incidence throughout the world. Although conventional modalities, like chemotherapy, have been applied for the inhibition of this cancer, they have not led to the suppression of lung cancer owing to their deficiencies. Thus, we developed a novel polylactic-co-glycolic acid (PLGA)-polyamidoamine G4 (PAMAM G4)-polycaprolactone (PCL) nanocarrier for efficient delivery of curcumin (Cur) to A549 lung cancer cells. The synthesized nanocarrier was characterized by applying analytical techniques, FT-IR, DLS, TEM, and TGA. Successful synthesis, nano-size diameter (40-80 nm), near neutral surface charge (8.0 mV), and high drug entrapment (11.5 %) were measured for the nanocarrier. Controlled (about 5 folds within first 2 h) and pH-sensitive (2-3 folds faster within first hours) Cur release observed for PLGA-PAMAM-PCL-Cur. Cell viability test (MTT assay) indicated the high capability of nanocarrier in suppression of A549 cancer cells (21 % viability after 24 h of treatment with 200 nM) while did not result in toxicity on MSC normal cells. The IC50 observed for 50 nM at 24 h of post-treatment in A549 cells. The qRT-PCR technique indicated inducing the expression of apoptotic genes (Caspase9 and Bax) by 6-8 folds and suppressing anti-apoptotic gene (Bcl2) by 7 folds. ROS considerably increased in cancer cells as well. This nanocarrier would be a promising drug delivery system against lung cancer.
Collapse
Affiliation(s)
- Yasir Qasim Almajidi
- Department of Pharmacy (pharmaceutics), Baghdad College of Medical Sciences, 10047, Baghdad, Iraq
- Department of Pharmaceutics, College of Pharmacy, Al-Nahrain university, 10072, Baghdad, Iraq
| | - Ali Q Jawad
- Department of Pharmaceutics, College of Pharmacy, Al-Nahrain university, 10072, Baghdad, Iraq
| | - Ahmed Abdulameer Albadri
- Department of Pharmacy (pharmaceutics), Baghdad College of Medical Sciences, 10047, Baghdad, Iraq
| |
Collapse
|
2
|
Vaidya K, Regan MS, Lin J, Houle J, Stopka SA, Agar NYR, Hammond PT, Boehnke N. Pooled nanoparticle screening using a chemical barcoding approach. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614746. [PMID: 39386478 PMCID: PMC11463557 DOI: 10.1101/2024.09.24.614746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
We report the development of a small molecule-based barcoding platform for pooled screening of nanoparticle delivery. Using aryl halide-based tags (halocodes), we achieve high-sensitivity detection via gas chromatography coupled with mass spectrometry or electron capture. This enables barcoding and tracking of nanoparticles with minimal halocode concentrations and without altering their physicochemical properties. To demonstrate the utility of our platform for pooled screening, we synthesized a halocoded library of polylactide-co-glycolide (PLGA) nanoparticles and quantified uptake in ovarian cancer cells in a pooled manner. Our findings correlate with conventional fluorescence-based assays. Additionally, we demonstrate the potential of halocodes for spatial mapping of nanoparticles using mass spectrometry imaging (MSI). Halocoding presents an accessible and modular nanoparticle screening platform capable of quantifying delivery of pooled nanocarrier libraries in a range of biological settings.
Collapse
|
3
|
Yang J, Zeng H, Luo Y, Chen Y, Wang M, Wu C, Hu P. Recent Applications of PLGA in Drug Delivery Systems. Polymers (Basel) 2024; 16:2606. [PMID: 39339068 PMCID: PMC11435547 DOI: 10.3390/polym16182606] [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: 06/21/2024] [Revised: 08/18/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Poly(lactic-co-glycolic acid) (PLGA) is a widely used biodegradable and biocompatible copolymer in drug delivery systems (DDSs). In this article, we highlight the critical physicochemical properties of PLGA, including its molecular weight, intrinsic viscosity, monomer ratio, blockiness, and end caps, that significantly influence drug release profiles and degradation times. This review also covers the extensive literature on the application of PLGA in delivering small-molecule drugs, proteins, peptides, antibiotics, and antiviral drugs. Furthermore, we discuss the role of PLGA-based DDSs in the treating various diseases, including cancer, neurological disorders, pain, and inflammation. The incorporation of drugs into PLGA nanoparticles and microspheres has been shown to enhance their therapeutic efficacy, reduce toxicity, and improve patient compliance. Overall, PLGA-based DDSs holds great promise for the advancement of the treatment and management of multiple chronic conditions.
Collapse
Affiliation(s)
- Jie Yang
- Department of Burns & Plastic Surgery, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou 510006, China
- College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Huiying Zeng
- College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Yusheng Luo
- International School, Jinan University, Guangzhou 510006, China
| | - Ying Chen
- Guangdong Institute for Drug Control, NMPA Key Laboratory for Quality Control and Evaluation of Pharmaceutical Excipients, Guangzhou 510660, China
| | - Miao Wang
- Guangdong Institute for Drug Control, NMPA Key Laboratory for Quality Control and Evaluation of Pharmaceutical Excipients, Guangzhou 510660, China
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Ping Hu
- Department of Burns & Plastic Surgery, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou 510006, China
- College of Pharmacy, Jinan University, Guangzhou 510006, China
| |
Collapse
|
4
|
Longobardi G, Moore TL, Conte C, Ungaro F, Satchi-Fainaro R, Quaglia F. Polyester nanoparticles delivering chemotherapeutics: Learning from the past and looking to the future to enhance their clinical impact in tumor therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1990. [PMID: 39217459 DOI: 10.1002/wnan.1990] [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: 05/29/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Polymeric nanoparticles (NPs), specifically those comprised of biodegradable and biocompatible polyesters, have been heralded as a game-changing drug delivery platform. In fact, poly(α-hydroxy acids) such as polylactide (PLA), poly(lactide-co-glycolide) (PLGA), and poly(ε-caprolactone) (PCL) have been heavily researched in the past three decades as the material basis of polymeric NPs for drug delivery applications. As materials, these polymers have found success in resorbable sutures, biodegradable implants, and even monolithic, biodegradable platforms for sustained release of therapeutics (e.g., proteins and small molecules) and diagnostics. Few fields have gained more attention in drug delivery through polymeric NPs than cancer therapy. However, the clinical translational of polymeric nanomedicines for treating solid tumors has not been congruent with the fervor or funding in this particular field of research. Here, we attempt to provide a comprehensive snapshot of polyester NPs in the context of chemotherapeutic delivery. This includes a preliminary exploration of the polymeric nanomedicine in the cancer research space. We examine the various processes for producing polyester NPs, including methods for surface-functionalization, and related challenges. After a detailed overview of the multiple factors involved with the delivery of NPs to solid tumors, the crosstalk between particle design and interactions with biological systems is discussed. Finally, we report state-of-the-art approaches toward effective delivery of NPs to tumors, aiming at identifying new research areas and re-evaluating the reasons why some research avenues have underdelivered. We hope our effort will contribute to a better understanding of the gap to fill and delineate the future research work needed to bring polyester-based NPs closer to clinical application. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
| | - Thomas Lee Moore
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Claudia Conte
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Francesca Ungaro
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Fabiana Quaglia
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
5
|
Hernández-Parra H, Cortés H, Romero-Montero A, Borbolla-Jiménez FV, Magaña JJ, Del Prado-Audelo ML, Florán B, Leyva-Gómez G. Polymeric nanoparticles decorated with fragmented chitosan as modulation systems for neuronal drug uptake. Carbohydr Polym 2024; 336:122121. [PMID: 38670753 DOI: 10.1016/j.carbpol.2024.122121] [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: 02/26/2024] [Revised: 03/22/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024]
Abstract
This study aimed to modify chitosan (CS) by gamma irradiation and use it as a surface coating of nanoparticles (NPs) fabricated of poly lactic-co-glycolic acid (PLGA) to create mostly biocompatible nanosystems that can transport drugs to neurons. Gamma irradiation produced irradiated CS (CSγ) with a very low molecular weight (15.2-19.2 kDa). Coating NPs-PLGA with CSγ caused significant changes in their Z potential, making it slightly positive (from -21.7 ± 2.8 mV to +7.1 ± 2.3 mV) and in their particle size (184.4 0.4 ± 7.9 nm to 211.9 ± 14.04 nm). However, these changes were more pronounced in NPs coated with non-irradiated CS (Z potential = +54.0 ± 1.43 mV, size = 348.1 ± 16.44 nm). NPs coated with CSγ presented lower cytotoxicity and similar internalization levels in SH-SY5Y neuronal cells than NPs coated with non-irradiated CS, suggesting higher biocompatibility. Highly biocompatible NPs are desirable as nanocarriers to deliver drugs to the brain, as they help maintain the structure and function of the blood-brain barrier. Therefore, the NPs developed in this study could be evaluated as drug-delivery systems for treating brain diseases.
Collapse
Affiliation(s)
- Hector Hernández-Parra
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, Mexico; Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, Mexico
| | - Alejandra Romero-Montero
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Fabiola V Borbolla-Jiménez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, Mexico
| | - Jonathan J Magaña
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, Mexico; Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Ciudad de México, 14380, Mexico.
| | | | - Benjamín Florán
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico.
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| |
Collapse
|
6
|
Brannon ER, Piegols LD, Cady G, Kupor D, Chu X, Guevara MV, Lima MR, Kanthi Y, Pinsky DJ, Uhrich KE, Eniola-Adefeso O. Polymerized Salicylic Acid Microparticles Reduce the Progression and Formation of Human Neutrophil Extracellular Traps (NET)s. Adv Healthc Mater 2024:e2400443. [PMID: 38898728 PMCID: PMC11628640 DOI: 10.1002/adhm.202400443] [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: 02/04/2024] [Revised: 06/02/2024] [Indexed: 06/21/2024]
Abstract
Neutrophils can contribute to inflammatory disease propagation via innate mechanisms intended for inflammation resolution. For example, neutrophil extracellular traps (NETs) are necessary for trapping pathogens but can contribute to clot formation and blood flow restriction, that is, ischemia. Currently, no therapeutics in the clinic directly target NETs despite the known involvement of NETs contributing to mortality and increased disease severity. Vascular-deployed particle-based therapeutics are a novel and robust alternative to traditional small-molecule drugs by enhancing drug delivery to cells of interest. This work designs a high-throughput assay to investigate the immunomodulatory behavior and functionality of salicylic acid-based polymer-based particle therapeutics against NETosis in human neutrophils. Briefly, this work finds that polymeric composition plays a role, and particle size can also influence rates of NETosis. Salicylate-based polymeric (Poly-SA) particles are found to functionally inhibit NETosis depending on the particle size and concentration exposed to neutrophils. This work demonstrates the high throughput method can help fast-track particle-based therapeutic optimization and design, more efficiently preparing this innovative therapeutics for the clinic.
Collapse
Affiliation(s)
- Emma R. Brannon
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, NCRC B28, Ann Arbor, MI, 48109, USA
| | - Logan D. Piegols
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, NCRC B28, Ann Arbor, MI, 48109, USA
| | - Gillian Cady
- Division of Cardiovascular Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Daniel Kupor
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, NCRC B28, Ann Arbor, MI, 48109, USA
| | - Xueqi Chu
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, NCRC B28, Ann Arbor, MI, 48109, USA
| | - M. Valentina Guevara
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, NCRC B28, Ann Arbor, MI, 48109, USA
| | - Mariana R.N. Lima
- Department of Chemistry, University of California Riverside, Riverside, CA
| | - Yogendra Kanthi
- Division of Cardiovascular Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
- Section of Vascular Thrombosis & Inflammation, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, 20892, USA
| | - David J. Pinsky
- Division of Cardiovascular Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kathryn E. Uhrich
- Department of Chemistry, University of California Riverside, Riverside, CA
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, NCRC B28, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| |
Collapse
|
7
|
Liu K, Wang L, Peng J, Lyu Y, Li Y, Duan D, Zhang W, Wei G, Li T, Niu Y, Zhao Y. Drug-Loaded Bacillus Calmette-Guérin Bacteria for Immuno-Chemo Combo Therapy in Bladder Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310735. [PMID: 38330363 DOI: 10.1002/adma.202310735] [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: 10/15/2023] [Revised: 02/02/2024] [Indexed: 02/10/2024]
Abstract
Intravesical Bacillus Calmette-Guérin (BCG) is a well-established strategy for managing high-risk nonmuscle-invasive bladder cancer (NMIBC); however, over half of patients still experience disease recurrence or progression. Although the combined intravesical instillation of various chemotherapeutic drugs is implemented in clinical trials to enhance the BCG therapy, the outcome is far from satisfying due to severe irritative effects and treatment intolerance at high doses. Therefore, it is adopted the "biotin-streptavidin strategy" to doxorubicin (DOX)-encapsulated nanoparticles within live BCG bacteria (DOX@BCG) to improve treatment outcomes. Adherence of BCG to the bladder epithelium helps precisely target DOX@BCG to the local tumor cells and simultaneously increases intratumoral transport of therapeutic drugs. DOX@BCG effectively inhibits cancer progression and prolongs the survival of rats/mice with orthotopic bladder cancer owing to synergism between BCG-immunotherapy, DOX-chemotherapy, and DOX-induced immunogenic tumor cell death; furthermore, it exhibits improved tolerance and biosafety, and establishes antitumor immunity in the tumor microenvironment. Therefore, the drug-loaded live BCG bacterial delivery system holds considerable potential for clinical translation in the intravesical treatment of bladder cancer.
Collapse
Affiliation(s)
- Kangkang Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Lining Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jing Peng
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yuanji Lyu
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yiming Li
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Dengyi Duan
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Wenyi Zhang
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Guojiang Wei
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Taipeng Li
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yuanjie Niu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yang Zhao
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| |
Collapse
|
8
|
Wu J, Wang X, Wang Y, Xun Z, Li S. Application of PLGA in Tumor Immunotherapy. Polymers (Basel) 2024; 16:1253. [PMID: 38732722 PMCID: PMC11085488 DOI: 10.3390/polym16091253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/24/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
Biodegradable polymers have been extensively researched in the field of biomedicine. Polylactic-co-glycolic acid (PLGA), a biodegradable polymer material, has been widely used in drug delivery systems and has shown great potential in various medical fields, including vaccines, tissue engineering such as bone regeneration and wound healing, and 3D printing. Cancer, a group of diseases with high mortality rates worldwide, has recently garnered significant attention in the field of immune therapy research. In recent years, there has been growing interest in the delivery function of PLGA in tumor immunotherapy. In tumor immunotherapy, PLGA can serve as a carrier to load antigens on its surface, thereby enhancing the immune system's ability to attack tumor cells. Additionally, PLGA can be used to formulate tumor vaccines and immunoadjuvants, thereby enhancing the efficacy of tumor immunotherapy. PLGA nanoparticles (NPs) can also enhance the effectiveness of tumor immunotherapy by regulating the activity and differentiation of immune cells, and by improving the expression and presentation of tumor antigens. Furthermore, due to the diverse physical properties and surface modifications of PLGA, it has a wider range of potential applications in tumor immunotherapy through the loading of various types of drugs or other innovative substances. We aim to highlight the recent advances and challenges of plga in the field of oncology therapy to stimulate further research and development of innovative PLGA-based approaches, and more effective and personalized cancer therapies.
Collapse
Affiliation(s)
- Jiashuai Wu
- Innovation Institute, China Medical University, Shenyang 110122, China; (J.W.); (X.W.)
| | - Xiaopeng Wang
- Innovation Institute, China Medical University, Shenyang 110122, China; (J.W.); (X.W.)
| | - Yunduan Wang
- School of Intelligent Medicine, China Medical University, Shenyang 110122, China;
| | - Zhe Xun
- Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Health Science Institute, China Medical University, Shenyang 110122, China
| | - Shuo Li
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang 110122, China
| |
Collapse
|
9
|
Dasanayake GS, Hamadani CM, Singh G, Kumar Misra S, Vashisth P, Sharp JS, Adhikari L, Baker GA, Tanner EEL. Imidazolium-based zwitterionic liquid-modified PEG-PLGA nanoparticles as a potential intravenous drug delivery carrier. NANOSCALE 2024; 16:5584-5600. [PMID: 38410026 PMCID: PMC11476077 DOI: 10.1039/d3nr06349f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Zwitterionic-based systems offer promise as next-generation drug delivery biomaterials capable of enhancing nanoparticle (NP) stimuli-responsiveness, biorecognition, and biocompatibility. Further, imidazole-functionalized amphiphilic zwitterions are able to readily bind to various biological macromolecules, enabling antifouling properties for enhanced drug delivery efficacy and bio-targeting. Herein, we describe structurally tuned zwitterionic imidazole-based ionic liquid (ZIL)-coated PEG-PLGA nanoparticles made with sonicated nanoprecipitation. Upon ZIL surface modification, the hydrodynamic radius increased by nearly 20 nm, and the surface charge significantly shifted closer to neutral. 1H NMR spectra suggests that the amount of ZIL on the nanoparticle surface is controlled by the structure of the ZIL and that the assembly occurs as a result of non-covalent interactions of ZIL-coated nanoparticle with the polymer surface. These nanoparticle-zwitterionic liquid (ZIL) constructs demonstrate selective affinity towards red blood cells in whole mouse blood and show relatively low human hemolysis at ∼5%. Additionally, we observe higher nanoparticle accumulation of ZIL-NPs compared with unmodified NP controls in human triple-negative breast cancer cells (MDA-MB-231). Furthermore, although the ZIL shows similar protein adsorption by SDS-PAGE, LC-MS/MS protein analysis data demonstrate a difference in the relative abundance and depletion of proteins in mouse and human serum. Hence, we show that ZIL-coated nanoparticles provide a new potential platform to enhance RBC-based drug delivery systems for cancer treatments.
Collapse
Affiliation(s)
- Gaya S Dasanayake
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA.
| | - Christine M Hamadani
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA.
| | - Gagandeep Singh
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA.
| | - Sandeep Kumar Misra
- Department of BioMolecular Sciences, University of Mississippi, University, MS 38677, USA
| | - Priyavrat Vashisth
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA.
| | - Joshua S Sharp
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA.
- Department of BioMolecular Sciences, University of Mississippi, University, MS 38677, USA
| | - Laxmi Adhikari
- Department of Chemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Gary A Baker
- Department of Chemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Eden E L Tanner
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA.
| |
Collapse
|
10
|
Ahmadi M, Ritter CA, von Woedtke T, Bekeschus S, Wende K. Package delivered: folate receptor-mediated transporters in cancer therapy and diagnosis. Chem Sci 2024; 15:1966-2006. [PMID: 38332833 PMCID: PMC10848714 DOI: 10.1039/d3sc05539f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/31/2023] [Indexed: 02/10/2024] Open
Abstract
Neoplasias pose a significant threat to aging society, underscoring the urgent need to overcome the limitations of traditional chemotherapy through pioneering strategies. Targeted drug delivery is an evolving frontier in cancer therapy, aiming to enhance treatment efficacy while mitigating undesirable side effects. One promising avenue utilizes cell membrane receptors like the folate receptor to guide drug transporters precisely to malignant cells. Based on the cellular folate receptor as a cancer cell hallmark, targeted nanocarriers and small molecule-drug conjugates have been developed that comprise different (bio) chemistries and/or mechanical properties with individual advantages and challenges. Such modern folic acid-conjugated stimuli-responsive drug transporters provide systemic drug delivery and controlled release, enabling reduced dosages, circumvention of drug resistance, and diminished adverse effects. Since the drug transporters' structure-based de novo design is increasingly relevant for precision cancer remediation and diagnosis, this review seeks to collect and debate the recent approaches to deliver therapeutics or diagnostics based on folic acid conjugated Trojan Horses and to facilitate the understanding of the relevant chemistry and biochemical pathways. Focusing exemplarily on brain and breast cancer, recent advances spanning 2017 to 2023 in conjugated nanocarriers and small molecule drug conjugates were considered, evaluating the chemical and biological aspects in order to improve accessibility to the field and to bridge chemical and biomedical points of view ultimately guiding future research in FR-targeted cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Mohsen Ahmadi
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
| | - Christoph A Ritter
- Institute of Pharmacy, Section Clinical Pharmacy, University of Greifswald Greifswald Germany
| | - Thomas von Woedtke
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
- Institute for Hygiene and Environmental Medicine, Greifswald University Medical Center Ferdinand-Sauerbruch-Straße 17475 Greifswald Germany
| | - Sander Bekeschus
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
- Clinic and Policlinic for Dermatology and Venereology, Rostock University Medical Center Strempelstr. 13 18057 Rostock Germany
| | - Kristian Wende
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
| |
Collapse
|
11
|
Toita R, Kitamura M, Tsuchiya A, Kang J, Kasahara S. Releasable, Immune-Instructive, Bioinspired Multilayer Coating Resists Implant-Induced Fibrosis while Accelerating Tissue Repair. Adv Healthc Mater 2024; 13:e2302611. [PMID: 38095751 PMCID: PMC11468989 DOI: 10.1002/adhm.202302611] [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: 08/10/2023] [Indexed: 12/21/2023]
Abstract
Implantable biomaterials trigger foreign body reactions (FBRs), which reduces the functional life of medical devices and prevents effective tissue regeneration. Although existing therapeutic approaches can circumvent collagen-rich fibrotic encapsulation secondary to FBRs, they disrupt native tissue repair. Herein, a new surface engineering strategy in which an apoptotic-mimetic, immunomodulatory, phosphatidylserine liposome (PSL) is released from an implant coating to induce the formation of a macrophage phenotype that mitigates FBRs and improves tissue healing is described. PSL-multilayers constructed on implant surfaces via the layer-by-layer method release PSLs over a 1-month period. In rat muscles, poly(etheretherketone) (PEEK), a nondegradable polymer implant model, induces FBRs with dense fibrotic scarring under an aberrant cellular profile that recruits high levels of inflammatory infiltrates, foreign body giant cells (FBGCs), scar-forming myofibroblasts, and inflammatory M1-like macrophages but negligible amounts of anti-inflammatory M2-like phenotypes. However, the PSL-multilayer coating markedly diminishes these detrimental signatures by shifting the macrophage phenotype. Unlike other therapeutics, PSL-multilayered coatings also stimulate muscle regeneration. This study demonstrates that PSL-multilayered coatings are effective in eliminating FBRs and promoting regeneration, hence offering potent and broad applications for implantable biomaterials.
Collapse
Affiliation(s)
- Riki Toita
- Biomedical Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)1‐8‐31 MidorigaokaIkedaOsaka563‐8577Japan
- AIST‐Osaka University Advanced Photonics and Biosensing Open Innovation LaboratoryAIST2‐1 YamadaokaSuitaOsaka565‐0871Japan
| | - Masahiro Kitamura
- Niterra Co., Ltd.2808 IwasakiKomakiAichi485–8510Japan
- NGK Spark Plug‐AIST Healthcare Materials Cooperative Research Laboratory2266–98 AnagahoraShimoshidami, Moriyama‐kuNagoyaAichi463–8560Japan
| | - Akira Tsuchiya
- Department of BiomaterialsFaculty of Dental ScienceKyushu University3‐1‐1 MaidashiHigashi‐kuFukuoka812–8582Japan
| | - Jeong‐Hun Kang
- Division of Biopharmaceutics and PharmacokineticsNational Cerebral and Cardiovascular Center Research Institute6‐1 Shinmachi, KishibeSuitaOsaka564–8565Japan
| | | |
Collapse
|
12
|
Gao X, Lang X, El Khoury E, Wei M, Qian N, Min W. Quantitative Label-Free Chemical Imaging of PLGA Nanoparticles in Cells and Tissues with Single-Particle Sensitivity. NANO LETTERS 2024; 24:1024-1033. [PMID: 38207237 DOI: 10.1021/acs.nanolett.3c04463] [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: 01/13/2024]
Abstract
Nanomedicine has brought significant advancements to healthcare by utilizing nanotechnology in medicine. Despite much promise, the further development of nanocarriers for clinical use has been hindered by a lack of understanding and visualization of nano-bio interactions. Conventional imaging methods have limitations in resolution, sensitivity, and specificity. This study introduces a label-free optical approach using stimulated Raman scattering (SRS) microscopy to image poly(lactic-co-glycolic acid) (PLGA) nanocarriers, the most widely used polymeric nanocarrier for delivery therapeutic agents, with single-particle sensitivity and quantification capabilities. A unique Raman peak was identified for PLGA ester, enabling generalized bio-orthogonal bond imaging. We demonstrated quantitative SRS imaging of PLGA nanocarriers across different biological systems from cells to animal tissues. This label-free imaging method provides a powerful tool for studying this prevalent nanocarrier and quantitatively visualizing their distribution, interaction, and clearance in vivo.
Collapse
Affiliation(s)
- Xin Gao
- Department of Chemistry, Columbia University, 3000 Broadway, New York, New York 10027, United States
| | - Xiaoqi Lang
- Department of Chemistry, Columbia University, 3000 Broadway, New York, New York 10027, United States
| | - Elsy El Khoury
- Department of Chemistry, Columbia University, 3000 Broadway, New York, New York 10027, United States
| | - Mian Wei
- Department of Chemistry, Columbia University, 3000 Broadway, New York, New York 10027, United States
| | - Naixin Qian
- Department of Chemistry, Columbia University, 3000 Broadway, New York, New York 10027, United States
| | - Wei Min
- Department of Chemistry, Columbia University, 3000 Broadway, New York, New York 10027, United States
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, New York 10027, United States
| |
Collapse
|
13
|
Chen J, Ren T, Xie L, Hu H, Li X, Maitusong M, Zhou X, Hu W, Xu D, Qian Y, Cheng S, Yu K, Wang JA, Liu X. Enhancing aortic valve drug delivery with PAR2-targeting magnetic nano-cargoes for calcification alleviation. Nat Commun 2024; 15:557. [PMID: 38228638 PMCID: PMC10792006 DOI: 10.1038/s41467-024-44726-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024] Open
Abstract
Calcific aortic valve disease is a prevalent cardiovascular disease with no available drugs capable of effectively preventing its progression. Hence, an efficient drug delivery system could serve as a valuable tool in drug screening and potentially enhance therapeutic efficacy. However, due to the rapid blood flow rate associated with aortic valve stenosis and the lack of specific markers, achieving targeted drug delivery for calcific aortic valve disease has proved to be challenging. Here we find that protease-activated-receptor 2 (PAR2) expression is up-regulated on the plasma membrane of osteogenically differentiated valvular interstitial cells. Accordingly, we develop a magnetic nanocarrier functionalized with PAR2-targeting hexapeptide for dual-active targeting drug delivery. We show that the nanocarriers effectively deliver XCT790-an anti-calcification drug-to the calcified aortic valve under extra magnetic field navigation. We demonstrate that the nano-cargoes consequently inhibit the osteogenic differentiation of valvular interstitial cells, and alleviate aortic valve calcification and stenosis in a high-fat diet-fed low-density lipoprotein receptor-deficient (Ldlr-/-) mouse model. This work combining PAR2- and magnetic-targeting presents an effective targeted drug delivery system for treating calcific aortic valve disease in a murine model, promising future clinical translation.
Collapse
Affiliation(s)
- Jinyong Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Tanchen Ren
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China.
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China.
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China.
| | - Lan Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Haochang Hu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Xu Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 200030, Shanghai, P.R. China
| | - Miribani Maitusong
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Xuhao Zhou
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Wangxing Hu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Dilin Xu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Yi Qian
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Si Cheng
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Kaixiang Yu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Jian An Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China.
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China.
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310053, P.R. China.
| | - Xianbao Liu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China.
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China.
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China.
| |
Collapse
|
14
|
Banerjee A, Kajol, Bajaj G, Singhal NK, Pathak RK. Synthetically Tunable Suprahybrid Nanoparticle Platform for the Efficacious Delivery of Therapeutics. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37927061 DOI: 10.1021/acsami.3c11626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
The discovery of lipid-hybrid nanosystems has offered potential solutions to various drug delivery and theranostic challenges. However, in many instances, the commonly used lipids and other components in these systems often pose challenges related to their solubility, physicochemical properties, immune compatibility, and limited synthetic tunability. In this work, we introduce a synthetically tunable supramolecular scaffold with amphiphilic characteristics based on the calix[4]arene macrocyclic system. We designed and synthesized two novel calix[4]arene-polyethylene glycol (PEG) conjugates, termed Cal-P1 and Cal-P2, and these were characterized utilizing a wide range of spectroscopic and analytical methods. The rational design of Cal-P1 and Cal-P2 demonstrates their utility in forming stable blended nanospheres with sustained drug release characteristics. The synergistic blending of PLGA and the calixarene scaffold (Cal-P1 and Cal-P2) in constructing long-lasting and controlled-release nanoparticles (NPs), which are optimized for encapsulating Nile Red dye, and their successful internalization and retention in HeLa cancer cells are demonstrated through in vitro assays. The potential of these NPs as sustained therapeutic carriers is investigated in vivo, showing improved retention compared to free dye with negligible toxicity. The successful design and construction of Cal-P1 and Cal-P2 nanosystems represent a new paradigm for addressing drug loading challenges, opening up opportunities for the development of highly efficient, synthetically tunable alternative adjuvants for drug encapsulation and delivery.
Collapse
Affiliation(s)
- Arka Banerjee
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Transit Campus: Industrial Training Institute (ITI) Berhampur Engineering School Road, Berhampur 760010, Odisha, India
| | - Kajol
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Transit Campus: Industrial Training Institute (ITI) Berhampur Engineering School Road, Berhampur 760010, Odisha, India
- Department of Biological Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Transit Campus: Industrial Training Institute (ITI) Berhampur Engineering School Road, Berhampur 760010, Odisha, India
| | - Geetika Bajaj
- National Agri-Food Biotechnology Institute (NABI), Sector-81, S.A.S. Nagar, Mohali, Punjab 140306, India
- Department of Biotechnology, Punjab University, Sector 25, Chandigarh 160014, India
| | - Nitin Kumar Singhal
- National Agri-Food Biotechnology Institute (NABI), Sector-81, S.A.S. Nagar, Mohali, Punjab 140306, India
| | - Rakesh Kumar Pathak
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Transit Campus: Industrial Training Institute (ITI) Berhampur Engineering School Road, Berhampur 760010, Odisha, India
| |
Collapse
|
15
|
Silvestri T, Grumetto L, Neri I, De Falco M, Graziano SF, Damiano S, Giaquinto D, Maruccio L, de Girolamo P, Villapiano F, Ciarcia R, Mayol L, Biondi M. Investigating the Effect of Surface Hydrophilicity on the Destiny of PLGA-Poloxamer Nanoparticles in an In Vivo Animal Model. Int J Mol Sci 2023; 24:14523. [PMID: 37833971 PMCID: PMC10572154 DOI: 10.3390/ijms241914523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/17/2023] [Accepted: 09/17/2023] [Indexed: 10/15/2023] Open
Abstract
This study aimed to examine the impact of different surface properties of poly(lactic-co-glycolic) acid (PLGA) nanoparticles (P NPs) and PLGA-Poloxamer nanoparticles (PP NPs) on their in vivo biodistribution. For this purpose, NPs were formulated via nanoprecipitation and loaded with diphenylhexatriene (DPH), a fluorescent dye. The obtained NPs underwent comprehensive characterization, encompassing their morphology, technological attributes, DPH release rate, and thermodynamic properties. The produced NPs were then administered to wild-type mice via intraperitoneal injection, and, at scheduled time intervals, the animals were euthanized. Blood samples, as well as the liver, lungs, and kidneys, were extracted for histological examination and biodistribution analysis. The findings of this investigation revealed that the presence of poloxamers led to smaller NP sizes and induced partial crystallinity in the NPs. The biodistribution and histological results from in vivo experiments evidenced that both, P and PP NPs, exhibited comparable concentrations in the bloodstream, while P NPs could not be detected in the other organs examined. Conversely, PP NPs were primarily sequestered by the lungs and, to a lesser extent, by the kidneys. Future research endeavors will focus on investigating the behavior of drug-loaded NPs in pathological animal models.
Collapse
Affiliation(s)
- Teresa Silvestri
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy;
| | - Lucia Grumetto
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (L.G.); (I.N.); (M.B.)
- National Institute of Biostructures and Biosystems (INBB), Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
| | - Ilaria Neri
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (L.G.); (I.N.); (M.B.)
- National Institute of Biostructures and Biosystems (INBB), Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
| | - Maria De Falco
- National Institute of Biostructures and Biosystems (INBB), Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
- Department of Biology, University Federico II of Naples, Via Cinthia 26, 80125 Naples, Italy;
| | - Sossio Fabio Graziano
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (L.G.); (I.N.); (M.B.)
| | - Sara Damiano
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino 1, 80137 Naples, Italy (D.G.); (L.M.)
| | - Daniela Giaquinto
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino 1, 80137 Naples, Italy (D.G.); (L.M.)
| | - Lucianna Maruccio
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino 1, 80137 Naples, Italy (D.G.); (L.M.)
| | - Paolo de Girolamo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino 1, 80137 Naples, Italy (D.G.); (L.M.)
| | - Fabrizio Villapiano
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (L.G.); (I.N.); (M.B.)
| | - Roberto Ciarcia
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino 1, 80137 Naples, Italy (D.G.); (L.M.)
| | - Laura Mayol
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), Piazzale Tecchio 80, 80125 Naples, Italy
| | - Marco Biondi
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy; (L.G.); (I.N.); (M.B.)
- Interdisciplinary Research Centre on Biomaterials (CRIB), Piazzale Tecchio 80, 80125 Naples, Italy
| |
Collapse
|
16
|
Hamadani CM, Dasanayake GS, Chism CM, Gorniak ME, Monroe WG, Merrell A, Pride MC, Heintz R, Wong K, Hossain M, Taylor G, Edgecomb SX, Jones D, Dhar J, Banka A, Singh G, Vashisth P, Randall J, Darlington DS, Everett J, Jarrett E, Werfel TA, Eniola-Adefeso O, Tanner EEL. Selective Blood Cell Hitchhiking in Whole Blood with Ionic Liquid-Coated PLGA Nanoparticles to Redirect Biodistribution After Intravenous Injection. RESEARCH SQUARE 2023:rs.3.rs-3146716. [PMID: 37502854 PMCID: PMC10371090 DOI: 10.21203/rs.3.rs-3146716/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Less than 5% of intravenously-injected nanoparticles (NPs) reach destined sites in the body due to opsonization and immune-based clearance in vascular circulation. By hitchhiking in situ onto specific blood components post-injection, NPs can selectively target tissue sites for unprecedentedly high drug delivery rates. Choline carboxylate ionic liquids (ILs) are biocompatible liquid salts <100X composed of bulky asymmetric cations and anions. This class of ILs has been previously shown to significantly extend circulation time and redirect biodistribution in BALB/c mice post-IV injection via hitchhiking on red blood cell (RBC) membranes. Herein, we synthesized & screened 60 choline carboxylic acid-based ILs to coat PLGA NPs and present the impact of structurally engineering the coordinated anion identity to selectively interface and hitchhike lymphocytes, monocytes, granulocytes, platelets, and RBCs in whole mouse blood for in situ targeted drug delivery. Furthermore, we find this nanoparticle platform to be biocompatible (non-cytotoxic), translate to human whole blood by resisting serum uptake and maintaining modest hitchhiking, and also significantly extend circulation retention over 24 hours in BALB/c healthy adult mice after IV injection. Because of their altered circulation profiles, we additionally observe dramatically different organ accumulation profiles compared to bare PLGA NPs. This study establishes an initial breakthrough platform for a modular and transformative targeting technology to hitchhike onto blood components with high efficacy and safety in the bloodstream post-IV administration.
Collapse
|
17
|
Narmani A, Jahedi R, Bakhshian-Dehkordi E, Ganji S, Nemati M, Ghahramani-Asl R, Moloudi K, Hosseini SM, Bagheri H, Kesharwani P, Khani A, Farhood B, Sahebkar A. Biomedical applications of PLGA nanoparticles in nanomedicine: advances in drug delivery systems and cancer therapy. Expert Opin Drug Deliv 2023; 20:937-954. [PMID: 37294853 DOI: 10.1080/17425247.2023.2223941] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 06/06/2023] [Indexed: 06/11/2023]
Abstract
INTRODUCTION During the last decades, the ever-increasing proportion of patients with cancer has been led to serious concerns worldwide. Therefore, the development and use of novel pharmaceuticals, like nanoparticles (NPs)-based drug delivery systems (DDSs), can be potentially effective in cancer therapy. AREA COVERED Poly lactic-co-glycolic acid (PLGA) NPs, as a kind of bioavailable, biocompatible, and biodegradable polymers, have approved by the Food and Drug Administration (FDA) for some biomedical and pharmaceutical applications. PLGA is comprised of lactic acid (LA) and glycolic acid (GA) and their ratio could be controlled during various syntheses and preparation approaches. LA/GA ratio determines the stability and degradation time of PLGA; lower content of GA results in fast degradation. There are several approaches for preparing PLGA NPs that can affect their various aspects, such as size, solubility, stability, drug loading, pharmacokinetics, and pharmacodynamics, and so on. EXPERT OPINION These NPs have indicated the controlled and sustained drug release in the cancer site and can use in passive and active (via surface modification) DDSs. This review aims to provide an overview of PLGA NPs, their preparation approach and physicochemical aspects, drug release mechanism and the cellular fate, DDSs for efficient cancer therapy, and status in the pharmaceutical industry and nanomedicine.
Collapse
Affiliation(s)
- Asghar Narmani
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Roghayyeh Jahedi
- Department of Plant Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Ehsan Bakhshian-Dehkordi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Saeid Ganji
- Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ruhollah Ghahramani-Asl
- Department of Medical Physics and Radiological Sciences, Faculty of Paramedicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Kave Moloudi
- Department of Radiology and Nuclear Medicine, Alley School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Mohammad Hosseini
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Hamed Bagheri
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Ali Khani
- Radiation Sciences Department, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Narmani A, Ganji S, Amirishoar M, Jahedi R, Kharazmi MS, Jafari SM. Smart chitosan-PLGA nanocarriers functionalized with surface folic acid ligands against lung cancer cells. Int J Biol Macromol 2023:125554. [PMID: 37356696 DOI: 10.1016/j.ijbiomac.2023.125554] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 05/15/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
Lung cancer is the second most prevalent and first killer cancer worldwide, and conventional approaches do not have enough ability to suppress it. Therefore, a novel targeted chitosan (CS)-poly lactic-co-glycolic acid (PLGA)-folic acid (FA) nanocarrier was developed for delivery of sorafenib (Sor) to lung cancer cells. The nanocarrier (CPSF) had a size of 30-40 nm with globular shapes. Surface charge and drug content of CPSF were ascertained at about 1.1 mV and 15 %, respectively. Controlled (4 % within 2 h) and pH-sensitive (18 % within 2 h at pH = 5.0) Sor release were observed for the nanocarrier. The MTT assay demonstrated a cell viability of 13 % after 24 h treatment with 400 nM CPSF in A549 cancer cells while it was 78 % in MSC normal cells. The qRT-PCR revealed >8 folds and 11 folds increase for Caspase9 and P53 genes after 5 h treatment with 100 nM (IC50) CPSF; but a reduction of 5 folds was observed for the Bcl2 gene. Besides, 57 % and 20 % apoptosis were attained in cell cycle arrest and apoptosis assays for CPSF, respectively. CPF indicated about 88 % internalization in cancer cells. These data prove that CPSF is a promising nanodelivery system for lung cancer suppression.
Collapse
Affiliation(s)
- Asghar Narmani
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, 1439957131 Tehran, Iran
| | - Saeid Ganji
- Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Maryam Amirishoar
- Department of Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roghayyeh Jahedi
- Department of Plant Biology, Faculty of Natural Sciences, University of Tabriz, 51666-16471 Tabriz, Iran
| | | | - Seid Mahdi Jafari
- Department of Food Materials and Process Design Engineering, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran.
| |
Collapse
|
19
|
Pacheco C, Baião A, Ding T, Cui W, Sarmento B. Recent advances in long-acting drug delivery systems for anticancer drug. Adv Drug Deliv Rev 2023; 194:114724. [PMID: 36746307 DOI: 10.1016/j.addr.2023.114724] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/20/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
The use of systemic anticancer chemotherapy is intrinsically limited by its toxicity. Whether dealing with small molecules or biopharmaceuticals, after systemic administration, small doses fail to reach effective intratumoral concentrations, while high doses with significant tumor inhibition effects may also drive the death of healthy cells, endangering the patients. Therefore, strategies based on drug delivery systems (DDSs) for avoiding the systemic toxicity have been designed. Due to their ability to protect drugs from early elimination and control drug release, DDSs can foster tumor exposure to anticancer therapeutics by extending their circulation time or steadily releasing drugs into the tumor sites. However, approval of tailored DDSs systems for clinical use is minimal as the safety and the in vivo activity still need to be ameliorated by manipulating their physicochemical characteristics. During the last few years, several strategies have been described to improve their safety, stability, and fine-tune pharmaceuticals release kinetics. Herein, we reviewed the main DDSs, namely polymeric conjugates, nano or microparticles, hydrogels, and microneedles, explored for long-acting anticancer treatments, highlighting recently proposed modifications and their potential advantages for different anticancer therapies. Additionally, important limitations of long-acting anticancer therapies and future technology directions were also covered.
Collapse
Affiliation(s)
- Catarina Pacheco
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Ana Baião
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Tao Ding
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal; Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China.
| |
Collapse
|
20
|
Smart nanosystem based on PLGA nanoparticles as potential candidate for photothermal therapy: Characterization and in vitro studies. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
|
21
|
Finbloom JA, Raghavan P, Kwon M, Kharbikar BN, Yu MA, Desai TA. Codelivery of synergistic antimicrobials with polyelectrolyte nanocomplexes to treat bacterial biofilms and lung infections. SCIENCE ADVANCES 2023; 9:eade8039. [PMID: 36662850 PMCID: PMC9858510 DOI: 10.1126/sciadv.ade8039] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
Bacterial biofilm infections, particularly those of Pseudomonas aeruginosa (PA), have high rates of antimicrobial tolerance and are commonly found in chronic wound and cystic fibrosis lung infections. Combination therapeutics that act synergistically can overcome antimicrobial tolerance; however, the delivery of multiple therapeutics at relevant dosages remains a challenge. We therefore developed a nanoscale drug carrier for antimicrobial codelivery by combining approaches from polyelectrolyte nanocomplex (NC) formation and layer-by-layer electrostatic self-assembly. This strategy led to NC drug carriers loaded with tobramycin antibiotics and antimicrobial silver nanoparticles (AgTob-NCs). AgTob-NCs displayed synergistic enhancements in antimicrobial activity against both planktonic and biofilm PA cultures, with positively charged NCs outperforming negatively charged formulations. NCs were evaluated in mouse models of lung infection, leading to reduced bacterial burden and improved survival outcomes. This approach therefore shows promise for nanoscale therapeutic codelivery to treat recalcitrant bacterial infections.
Collapse
Affiliation(s)
- Joel A. Finbloom
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Preethi Raghavan
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Michael Kwon
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Bhushan N. Kharbikar
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Michelle A. Yu
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Tejal A. Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- School of Engineering, Brown University, Providence, RI, USA
| |
Collapse
|
22
|
Nanotechnology-Based RNA Vaccines: Fundamentals, Advantages and Challenges. Pharmaceutics 2023; 15:pharmaceutics15010194. [PMID: 36678823 PMCID: PMC9864317 DOI: 10.3390/pharmaceutics15010194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023] Open
Abstract
Over the past decades, many drugs based on the use of nanotechnology and nucleic acids have been developed. However, until recently, most of them remained at the stage of pre-clinical development and testing and did not find their way to the clinic. In our opinion, the main reason for this situation lies in the enormous complexity of the development and industrial production of such formulations leading to their high cost. The development of nanotechnology-based drugs requires the participation of scientists from many and completely different specialties including Pharmaceutical Sciences, Medicine, Engineering, Drug Delivery, Chemistry, Molecular Biology, Physiology and so on. Nevertheless, emergence of coronavirus and new vaccines based on nanotechnology has shown the high efficiency of this approach. Effective development of vaccines based on the use of nucleic acids and nanomedicine requires an understanding of a wide range of principles including mechanisms of immune responses, nucleic acid functions, nanotechnology and vaccinations. In this regard, the purpose of the current review is to recall the basic principles of the work of the immune system, vaccination, nanotechnology and drug delivery in terms of the development and production of vaccines based on both nanotechnology and the use of nucleic acids.
Collapse
|
23
|
Ghosh A, Rajdev B, Parihar N, Ponneganti S, Das P, Naidu VGM, Krishnanand P R, USN M, Kumar J, Pemmaraju DB. Bio-nanoconjugates of lithocholic acid/IR 780 for ROS-mediated apoptosis and optoacoustic imaging applications in breast cancer. Colloids Surf B Biointerfaces 2022; 221:113023. [DOI: 10.1016/j.colsurfb.2022.113023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/30/2022] [Accepted: 11/13/2022] [Indexed: 11/16/2022]
|
24
|
Lopez-Cazares G, Eniola-Adefeso O. Dual Coating of Chitosan and Albumin Negates the Protein Corona-Induced Reduced Vascular Adhesion of Targeted PLGA Microparticles in Human Blood. Pharmaceutics 2022; 14:pharmaceutics14051018. [PMID: 35631604 PMCID: PMC9143524 DOI: 10.3390/pharmaceutics14051018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/30/2022] [Accepted: 05/06/2022] [Indexed: 12/13/2022] Open
Abstract
Vascular-targeted carriers (VTCs) have the potential to localize therapeutics and imaging agents to inflamed, diseased sites. Poly (lactic-co-glycolic acid) (PLGA) is a negatively charged copolymer commonly used to construct VTCs due to its biodegradability and FDA approval. Unfortunately, PLGA VTCs experienced reduced adhesion to inflamed endothelium in the presence of human plasma proteins. In this study, PLGA microparticles were coated with chitosan (CS), human serum albumin (HSA), or both (HSA-CS) to improve adhesion. The binding of sialyl Lewis A (a ligand for E-selectin)-targeted PLGA, HSA-PLGA, CSPLGA, and HSA-CSPLGA to activated endothelial cells was evaluated in red blood cells in buffer or plasma flow conditions. PLGA VTCs with HSA-only coating showed improvement and experienced 35–52% adhesion in plasma compared to plasma-free buffer conditions across all shear rates. PLGA VTCs with dual coating—CS and HSA—maintained 80% of their adhesion after exposure to plasma at low and intermediate shears and ≈50% at high shear. Notably, the protein corona characterization showed increases at the 75 and 150 kDa band intensities for HSA-PLGA and HSA-CSPLGA, which could correlate to histidine-rich glycoprotein and immunoglobulin G. The changes in protein corona on HSA-coated particles seem to positively influence particle binding, emphasizing the importance of understanding plasma protein–particle interactions.
Collapse
Affiliation(s)
- Genesis Lopez-Cazares
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence: ; Tel.: +1-734-936-0856
| |
Collapse
|
25
|
Davis G, Kurse A, Agarwal A, Sheikh-Hamad D, Kumar MNVR. Nano-encapsulation strategies to circumvent drug-induced kidney injury and targeted nanomedicines to treat kidney diseases. CURRENT OPINION IN TOXICOLOGY 2022. [DOI: 10.1016/j.cotox.2022.100346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|