1
|
Imtiaz S, Ferdous UT, Nizela A, Hasan A, Shakoor A, Zia AW, Uddin S. Mechanistic study of cancer drug delivery: Current techniques, limitations, and future prospects. Eur J Med Chem 2025; 290:117535. [PMID: 40132495 DOI: 10.1016/j.ejmech.2025.117535] [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: 02/26/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Cancer drug delivery remains a critical challenge with systemic toxicity, poor drug bioavailability, and a lack of effective targeting. Overcoming these barriers is essential for improving treatment efficacy and patient outcomes. This review discusses current drug delivery techniques that reshape cancer therapy by offering precise, controlled-release tailored to tumor-specific features. Innovations in nanotechnology, immunotherapy, and gene therapy enable interventions at molecular and cellular levels. Radiomics and pathomics integrate high-dimensional data to optimize diagnostics and treatment planning. Combination therapy addresses the complexities of tumor heterogeneity by synergizing multiple agents within a single therapeutic framework, while peptide-drug conjugates enhance specificity and potency. Hydrogel-based systems and microneedle arrays offer localized, sustained release, significantly improving therapeutic outcomes. However, clinical translation of these advancements faces significant barriers such as drug resistance, off-target effects, scalability, cost, and ethical concerns. Moreover, regulatory complexities and the economic feasibility of these therapies highlight the need for innovative frameworks to make them accessible globally. Therefore, there is a need for innovation in gene and cell therapy, next-generation drug delivery platforms, and personalized medicine. This review focuses on recent advancements in drug delivery techniques over the past decade, evaluating their limitations and exploring potential future directions for transforming cancer treatment.
Collapse
Affiliation(s)
- Saiqa Imtiaz
- Department of Bioengineering, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia
| | - Umme Tamanna Ferdous
- Center for Biosystems and Machines, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia
| | - Alexis Nizela
- Department of Bioengineering, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia; Center for Membranes and Water Security, King Fahd University of Petroleum and Minerals, Dhahran, 31261, Saudi Arabia
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha, 2713, Qatar; Biomedical Research Center, Qatar University, Doha, 2713, Qatar
| | - Adnan Shakoor
- Center for Biosystems and Machines, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia; Department of Control & Instrumentation Engineering, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia
| | - Abdul Wasy Zia
- Institute of Mechanical, Process, and Energy Engineering (IMPEE), School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, United Kingdom.
| | - Shihab Uddin
- Department of Bioengineering, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia; Center for Biosystems and Machines, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia.
| |
Collapse
|
2
|
Nasrine A, Mohanto S, Narayana S, Ahmed MG. Enhanced pharmacokinetic approach for anastrozole via macromolecule-based silk fibroin nanoparticles incorporated in situ injectables for oestrogen-positive breast cancer therapy. J Drug Target 2025; 33:793-803. [PMID: 39754343 DOI: 10.1080/1061186x.2024.2449486] [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: 10/05/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/06/2025]
Abstract
Breast cancer (BC) is a substantial reason for cancer-related mortality among women across the globe. Anastrozole (ANS) is an effective orally administered hormonal therapy for oestrogen-positive (ER+) BC treatment. However, several side effects and pharmacokinetic limitations restricted the uses of ANS in BC therapy. Therefore, this investigation developed an in situ gelling injectable-loaded silk fibroin (SF)-ANS NPs, which offers sustained drug release and improved pharmacokinetic properties compared to conventional oral formulations at the targeted site. The optimised in situ gel (ISG) incorporated SF-ANS-NPs were developed, and the pharmacokinetic parameters were accessed in subcutaneous administration of NMU-induced Wistar albino rats. The results demonstrated that SF-ANS-NP-ISG exhibited a significantly higher Cmax, Tmax, and AUC compared to pure ANS suspension. In addition, tumour multiplicity (1.40 ± 0.66), tumour latency (75 ± 9.2 days), and incidence rate (90 ± 2.1%) were recorded, and post-treatment analysis reported a marked reduction in tumour volume and weight compared to positive control within 90 days of a single dose. The SF-ANS-NP-ISG treated group's histopathological assessment indicated a low-grade carcinoma, reduced epithelial hyperplasia, and haemorrhage in mammary tumour tissues compared to positive control. Thus, the SF-ANS-NPs-ISG investigated to overcome the pharmacokinetic limitations of ANS further exhibited targeted delivery and bioavailability compared to conventional dosage forms.
Collapse
Affiliation(s)
- Arfa Nasrine
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Bangalore, India
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Soumya Narayana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| |
Collapse
|
3
|
Beyaz H, Kavaz D, Rizaner N. Chitosan nanoparticle encapsulation of thymus capitatus essential oil: in vitro release, antioxidant, antibacterial activity and cytotoxicity in MDA-MB-231 cells. Pharm Dev Technol 2025:1-15. [PMID: 40163347 DOI: 10.1080/10837450.2025.2487255] [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: 11/28/2024] [Revised: 03/07/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Thymus capitatus (Th. Ca) is known to treat mouth ulcers and respiratory infections in Cyprus. However, antioxidant, antibacterial, and cytotoxic potential of Th. Ca. EO on MDA-MB-231 cells and its' encapsulation into nanoparticles has not been well studied. Therefore, we aimed to analyze the antioxidant, antibacterial, cytotoxic potential, loading efficiency, and in vitro release profile of both Th. Ca. EO and Chitosan Nanoparticle (Ch. Np) - Th. Ca. EO. GC-MS analysis revealed 53.97% carvacrol, 14.53% borneol, and 12.09% sabinene presence in EO. The loading efficiency of Th. Ca. EO into Ch. Np. was calculated as 35.27% and the in vitro release profile reached a maximum of 68% in pH 7 for two weeks. The Minimum Inhibitory Concentration (MIC) assay showed that E. coli had an MIC50 of 0.3215 mg/ml while B. subtilis had an MIC50 of 0.5304 mg/ml. The antioxidant activity of the EO was assessed by performing a DPPH assay with an IC50 = 440 μg/ml. Trypan Blue Assay revealed that 60 µg/ml Th. Ca. EO significantly reduced the cell viability of MDA-MB-231 cells by 10.7% at 48h and 20.06% at 72h. Overall, Ch. Np. - Th. Ca. EO has shown a promising formulation for the pharmaceutical industry.
Collapse
Affiliation(s)
- Huseyin Beyaz
- Bioengineering Department, Faculty of Engineering, Cyprus International University, Turkey
| | - Doga Kavaz
- Bioengineering Department, Faculty of Engineering, Cyprus International University, Turkey
- Biotechnology Research Centre, Cyprus International University, Nicosia, Turkey
| | - Nahit Rizaner
- Bioengineering Department, Faculty of Engineering, Cyprus International University, Turkey
- Biotechnology Research Centre, Cyprus International University, Nicosia, Turkey
| |
Collapse
|
4
|
Baek EJ, Nguyen HD, Ngo HV, Gil MC, Lee BJ. Long-term controlled release with reduced initial burst release utilizing calcium ion-triggering nanoaggregates of pasireotide-loaded fattigated albumin nanoparticles. Int J Pharm 2025; 673:125401. [PMID: 40010529 DOI: 10.1016/j.ijpharm.2025.125401] [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: 12/16/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
The aim of this study was to investigate the long-term controlled release of peptide-loaded fattigated albumin nanoparticles via calcium ion-triggering nanoaggregation with minimal initial burst release. Fattigated albumin nanoparticles were prepared via sonication by the self-assembly of human serum albumin (HSA)-oleic acid conjugates (AOC) with three different substitution ratios of oleic acid (OA) to modulate hydrophobicity. Then, pasireotide pamoate (PAS) as a model peptide was encapsulated into the hydrophobic core of HSA-OA nanoparticles (PAS-AONs). The critical micelle concentration of AOC decreased as OA substitution ratio increased. The loading efficiency of PAS increased owing to the strong hydrophobic-hydrophobic interactions between PAS and the hydrophobic block in the AONs. The release rate was also delayed, whereas the initial burst release was minimized, as the hydrophobicity of AOC increased. Interestingly, calcium ions triggered the formation of nanoaggregates of negatively charged PAS-AONs via electrostatic interactions, resulting in a further decrease in the release rate for one month via a reduced surface area while minimizing the initial burst release in a calcium ion concentration-dependent manner. The modulation of OA substitutions and calcium ion concentration of AONs could provide the potential for long-term delivery of peptide drugs while minimizing the initial huge burst release and controlling the release rate.
Collapse
Affiliation(s)
- Eun Jin Baek
- Department of Pharmacy, College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Hy Dinh Nguyen
- Department of Pharmacy, College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Hai Van Ngo
- Department of Pharmacy, College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | | | - Beom-Jin Lee
- Department of Pharmacy, College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
5
|
Muhammad AM, Ismail A, Chong PP, Yap WH, Muhamad A, Alitheen NB, Kam A, Loo S, Lee KW. Skin-penetrating peptides (SKPs): Enhancing skin permeation for transdermal delivery of pharmaceuticals and cosmetic compounds. Int J Pharm 2025; 672:125339. [PMID: 39947363 DOI: 10.1016/j.ijpharm.2025.125339] [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: 11/19/2024] [Revised: 01/16/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Skin-penetrating peptides (SKPs) are emerging as a promising class of permeation enhancers that can facilitate macromolecule delivery across the skin. Although their pharmaceutical applications are under extensive study, SKPs are crucial for enhancing skin permeability, enabling larger molecules to penetrate the stratum corneum. This review explores the transformative role of SKPs in non-invasive transdermal drug delivery. Drawing from an extensive collection of literature, it provides insights into the current usage and application of SKPs as tools to enhance skin permeability and facilitate the delivery of larger molecules. Additionally, it highlights the opportunities, challenges, and future directions for SKP applications in transdermal drug delivery.
Collapse
Affiliation(s)
- Ameerah Montree Muhammad
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Alif Ismail
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Pei Pei Chong
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia; Digital Health and Medical Advancement Impact Lab, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Wei Hsum Yap
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia; Food Security and Nutrition Impact Lab, Taylor's University, Subang Jaya 47500 Selangor, Malaysia
| | - Azira Muhamad
- Malaysia Genome and Vaccine Institute, National Institutes of Biotechnology Malaysia Kajang Selangor Malaysia
| | - Noorjahan Banu Alitheen
- Faculty of Biotechnology & Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Antony Kam
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Shining Loo
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Khai Wooi Lee
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia; Digital Health and Medical Advancement Impact Lab, Taylor's University, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
6
|
Kaga E, Kaga S, Altunbas K, Okumus N. Inhalable Nano Formulation of Cabazitaxel: A Comparative Study with Intravenous Route. Macromol Biosci 2025:e2400567. [PMID: 39888152 DOI: 10.1002/mabi.202400567] [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: 11/19/2024] [Revised: 12/25/2024] [Indexed: 02/01/2025]
Abstract
Chemotherapy is generally given by intravenous (IV) administration which provides higher bioavailability than other systemic routes. However, in the case of lung cancer, the pulmonary (INH) route is the other choice for inhalable formulations. In the study, biochemical and histological parameters of Cabazitaxel (CBZ) free (2 mg kg-1) and nanoparticle (NP) (2 mg kg-1 CBZ equivalent) formulations are investigated after IV and INH administration in rats. The nanoformulation of CBZ is obtained using PEGylated polystyrene (PEG-PST) nanoparticles obtained by PISA. While a nose and head-only device is used for INH administration, a jugular vein is used as the IV route. Blood samples (blank, 24 h, and 48 h) are collected via carotid artery cannulas without handling in metabolism cages. According to biochemical parameters, free CBZ formulation applied via IV or INH route shows higher systemic toxicity. On the other hand, the nanoformulation of CBZ showed no signs of toxicity in both IV or INH routes. Higher and longer retention is observed in the case of inhaled nanoformulation. Histological analysis showed higher alveolar macrophage migration for inhaled nanoformulation due to enhanced retention. Results showed that nanotechnology and the lung defense system gave the advantage to end up with an inhalable nanomedicine formulation for lung cancer.
Collapse
Affiliation(s)
- Elif Kaga
- Department of Medical Services and Techniques, Afyonkarahisar Health Sciences University, Afyonkarahisar, 03030, Türkiye
| | - Sadik Kaga
- Department of Biomedical Engineering, Afyon Kocatepe University, Afyonkarahisar, 03200, Türkiye
| | - Korhan Altunbas
- Department of Histology and Embryology, Afyon Kocatepe University, Afyonkarahisar, 03200, Türkiye
| | - Nurullah Okumus
- Department of Pediatrics, Afyonkarahisar Health Sciences University, Afyonkarahisar, 03030, Türkiye
| |
Collapse
|
7
|
Wang L, Wu X, Wang X, Dong M, Zhang H, Zhao P. Targeting CHEK1: Ginsenosides-Rh2 and Cu2O@G-Rh2 nanoparticles in thyroid cancer. Cell Biol Toxicol 2025; 41:30. [PMID: 39808342 PMCID: PMC11732901 DOI: 10.1007/s10565-024-09961-7] [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] [Received: 06/21/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025]
Abstract
Thyroid cancer (THCA) is an increasingly common malignant tumor of the endocrine system, with its incidence rising steadily in recent years. For patients who experience recurrence or metastasis, treatment options are relatively limited, and the prognosis is poor. Therefore, exploring new therapeutic strategies has become particularly urgent. This study confirmed that effective suppression of THCA cell proliferation and stimulation of apoptosis can be achieved through the application of Ginsenosides-Rh2. Through network pharmacology screening, the molecular target of Ginsenosides-Rh2 in THCA was identified as CHEK1, and its inhibitory effect was confirmed by downregulating CHEK1 protein expression. Furthermore, demonstrations conducted both in vitro and in vivo showcased that delivering Ginsenosides-Rh2 using nanoparticle carriers significantly reduced cell viability by approximately 50%, regulated DNA damage levels, apoptosis-related protein expression, and cell cycle control. The IC50 of the nanoparticle formulation was determined (B-CPAP IC50 = 88.24 μM), TPC IC50 = 79.52 μM). This study confirmed that Cu2O@G-Rh2 is effective in suppressing tumors and exhibits a significant inhibitory effect on tumor recurrence and metastasis while maintaining good safety. Cu2O@G-Rh2 nanoparticles possess excellent stability and anti-tumor efficacy. This research offers new perspectives for the treatment of THCA and demonstrates potential clinical applications.
Collapse
Affiliation(s)
- Lidong Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Xin Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - XinLu Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Meng Dong
- Department of Ultrasound, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Hao Zhang
- Department of Ultrasound, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China.
| | - Pengfei Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
8
|
Muhammad FA, Altalbawy FMA, Mandaliya V, Saraswat SK, Rekha MM, Aulakh D, Chahar M, Mahdi MS, Jaber MA, Alhadrawi M. Targeting breast tumor extracellular matrix and stroma utilizing nanoparticles. Clin Transl Oncol 2024:10.1007/s12094-024-03793-x. [PMID: 39692807 DOI: 10.1007/s12094-024-03793-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024]
Abstract
Breast cancer is a complicated malignancy and is known as the most common cancer in women. Considerable experiments have been devoted to explore the basic impacts of the tumor stroma, particularly the extracellular matrix (ECM) and stromal components, on tumor growth and resistance to treatment. ECM is made up of an intricate system of proteins, glycosaminoglycans, and proteoglycans, and maintains structural support and controls key signaling pathways involved in breast tumors. ECM can block different drugs such as chemotherapy and immunotherapy drugs from entering the tumor stroma. Furthermore, the stromal elements, such as cancer-associated fibroblasts (CAFs), immune cells, and blood vessels, have crucial impacts on tumor development and therapeutic resistance. Recently, promising outcomes have been achieved in using nanotechnology for delivering drugs to tumor stroma and crossing ECM in breast malignancies. Nanoparticles have various benefits for targeting the breast tumor stroma, such as improved permeability and retention, extended circulation time, and the ability to actively target the area. This review covers the latest developments in nanoparticle therapies that focus on breast tumor ECM and stroma. We will explore different approaches using nanoparticles to target the delivery of anticancer drugs like chemotherapy, small molecule drugs, various antitumor products, and other specific synthetic therapeutic agents to the breast tumor stroma. Furthermore, we will investigate the utilization of nanoparticles in altering the stromal elements, such as reprogramming CAFs and immune cells, and also remodeling ECM.
Collapse
Affiliation(s)
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, 12613, Egypt.
| | - Viralkumar Mandaliya
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | | | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Damanjeet Aulakh
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology Chitkara University, Rajpura, Punjab, 140401, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering and Technology, NIMS University Rajasthan, Jaipur, India
| | | | | | - Merwa Alhadrawi
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
9
|
Cimino C, Zingale E, Bonaccorso A, Musumeci T, Carbone C, Pignatello R. From Preformulative Design to In Vivo Tests: A Complex Path of Requisites and Studies for Nanoparticle Ocular Application. Part 1: Design, Characterization, and Preliminary In Vitro Studies. Mol Pharm 2024; 21:6034-6061. [PMID: 39441703 DOI: 10.1021/acs.molpharmaceut.4c00554] [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: 10/25/2024]
Abstract
Ocular pathologies are widely diffused worldwide, and their effective treatment, combined with a high patient compliance, is sometimes challenging to achieve due to the barriers of the eye; in this context, the use of nanoparticles for topical ophthalmic application could represent a successful strategy. Aiming to develop nanoplatforms with potential clinical applications, great attention has to be paid to their features, in relation to the route of administration and to the pharmacopoeial requirements. This review (part 1) thus embraces the preliminary steps of nanoparticle development and characterization. At the beginning, the main barriers of the eye and the different administration routes are resumed, followed by a general description of the advantages of the employment of nanoparticles for ocular topical administration. Subsequently, the preformulative steps are discussed, deepening the choice of raw materials and determining the quantitative composition. Then, a detailed report of the physicochemical and technological characterization of nanoparticles is presented, analyzing the most relevant tests that should be performed on nanoparticles to verify their properties and the requisites (both mandatory and suggested) demanded by regulatory agencies. In conclusion, some preliminary noncellular in vitro evaluation methods are described. Studies from in vitro cellular assays to in vivo tests will be discussed in a separate (part 2) review paper. Hence, this overview aims to offer a comprehensive tool to guide researchers in the choice of the most relevant studies to develop a nanoplatform for ophthalmic drug administration.
Collapse
Affiliation(s)
- Cinzia Cimino
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| | - Elide Zingale
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| | - Angela Bonaccorso
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| | - Teresa Musumeci
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| | - Claudia Carbone
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| | - Rosario Pignatello
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95124 Catania, Italy
| |
Collapse
|
10
|
Mishra S, Shelar SB, Rout S, Hassan PA, Barick KC, Agarwal N. Enhanced Singlet Oxygen Generation in Aggregates of Naphthalene-Fused BODIPY and Its Application in Photodynamic Therapy. ACS APPLIED BIO MATERIALS 2024; 7:7207-7218. [PMID: 39445398 DOI: 10.1021/acsabm.4c00804] [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: 10/25/2024]
Abstract
Several reports are available on aggregation-induced emission and its applications in biomedical imaging and other material sciences. However, enhancement of singlet oxygen generation in nanoaggregates is rarely reported. Here, we report the synthesis of Naph-BODIPY Br2, which absorbs at 661 nm (monomer) with a high molar absorption coefficient. The presence of bromine promotes intersystem crossing, thereby enhancing the singlet oxygen quantum yield (ΦΔ ∼ 0.50 in methanol). In order to increase hydrophilicity, we developed Naph-BODIPY Br2 nanoaggregates (∼100 nm), which demonstrated aggregation-induced properties and exhibited a bathochromic shift with an absorption maximum at 757 nm. The bathochromic shift in the UV-vis spectra due to aggregation is corroborated by TD-DFT analysis. The computational data also confirm the presence of a low-lying triplet state, which enhances the generation of singlet oxygen, making it effective for photodynamic therapy. These aggregates showed excellent singlet oxygen generation in aqueous media, compared to their monomeric form and standard methylene blue. Their hydrophilic nature and high singlet oxygen generation enabled significant phototoxicity against human carcinoma cells with IC50 values of 4.06 ± 0.01 and 4.09 ± 0.1 μM, respectively, for MCF-7 and A549 cells upon 5 min exposure to light. Moreover, their phototoxicity further increases with an increasing exposure time of light for both cell lines. Notably, Naph-BODIPY Br2 nanoaggregates exhibited nearly zero dark cell toxicity and effectively induced apoptosis in cancer cells upon light activation, highlighting their potential as powerful photosensitizers for photodynamic cancer therapy.
Collapse
Affiliation(s)
- Sneha Mishra
- School of Chemical Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Kalina, Santacruz (E), Mumbai 400098, India
| | | | - Saiprakash Rout
- School of Chemical Sciences, National Institute of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni, Khurda752050,India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Puthusserickal A Hassan
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - K C Barick
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Neeraj Agarwal
- School of Chemical Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Kalina, Santacruz (E), Mumbai 400098, India
| |
Collapse
|
11
|
Wilar G, Suhandi C, Wathoni N, Fukunaga K, Kawahata I. Nanoparticle-Based Drug Delivery Systems Enhance Treatment of Cognitive Defects. Int J Nanomedicine 2024; 19:11357-11378. [PMID: 39524925 PMCID: PMC11550695 DOI: 10.2147/ijn.s484838] [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: 06/29/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Nanoparticle-based drug delivery presents a promising solution in enhancing therapies for neurological diseases, particularly cognitive impairment. These nanoparticles address challenges related to the physicochemical profiles of drugs that hinder their delivery to the central nervous system (CNS). Benefits include improved solubility due to particle size reduction, enhanced drug penetration across the blood-brain barrier (BBB), and sustained release mechanisms suitable for long-term therapy. Successful application of nanoparticle delivery systems requires careful consideration of their characteristics tailored for CNS delivery, encompassing particle size and distribution, surface charge and morphology, loading capacity, and drug release kinetics. Literature review reveals three main types of nanoparticles developed for cognitive function enhancement: polymeric nanoparticles, lipid-based nanoparticles, and metallic or inorganic nanoparticles. Each type and its production methods possess distinct advantages and limitations. Further modifications such as coating agents or ligand conjugation have been explored to enhance their brain cell uptake. Evidence supporting their development shows improved efficacy outcomes, evidenced by enhanced cognitive function assessments, modulation of pro-oxidant markers, and anti-inflammatory activities. Despite these advancements, clinical trials validating the efficacy of nanoparticle systems in treating cognitive defects are lacking. Therefore, these findings underscore the need for researchers to expedite clinical testing to provide robust evidence of the potential of nanoparticle-based drug delivery systems.
Collapse
Affiliation(s)
- Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
| |
Collapse
|
12
|
Elhassan E, Omolo CA, Gafar MA, Kiruri LW, Ibrahim UH, Ismail EA, Devnarain N, Govender T. Disease-Inspired Design of Biomimetic Tannic Acid-Based Hybrid Nanocarriers for Enhancing the Treatment of Bacterial-Induced Sepsis. Mol Pharm 2024; 21:4924-4946. [PMID: 39214595 DOI: 10.1021/acs.molpharmaceut.4c00048] [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: 09/04/2024]
Abstract
This study explored the development of novel biomimetic tannic acid-based hybrid nanocarriers (HNs) for targeted delivery of ciprofloxacin (CIP-loaded TAH-NPs) against bacterial-induced sepsis. The prepared CIP-loaded TAH-NPs exhibited appropriate physicochemical characteristics and demonstrated biocompatibility and nonhemolytic properties. Computational simulations and microscale thermophoresis studies validated the strong binding affinity of tannic acid (TA) and its nanoformulation to human Toll-like receptor 4, surpassing that of the natural substrate lipopolysaccharide (LPS), suggesting a potential competitive inhibition against LPS-induced inflammatory responses. CIP released from TAH-NPs displayed a sustained release profile over 72 h. The in vitro antibacterial activity studies revealed that CIP-loaded TAH-NPs exhibited enhanced antibacterial efficacy and efflux pump inhibitory activity. Specifically, they showed a 3-fold increase in biofilm eradication activity against MRSA and a 2-fold increase against P. aeruginosa compared to bare CIP. Time-killing assays demonstrated complete bacterial clearance within 8 h of treatment with CIP-loaded TAH-NPs. In vitro DPPH scavenging and anti-inflammatory investigations confirmed the ability of the prepared hybrid nanosystem to neutralize reactive oxygen species (ROS) and modulate LPS-induced inflammatory responses. Collectively, these results suggest that CIP-loaded TAH-NPs may serve as an innovative nanocarrier for the effective and targeted delivery of antibiotics against bacterial-induced sepsis.
Collapse
Affiliation(s)
- Eman Elhassan
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, P.O. Box 14634-00800, Nairobi 00800, Kenya
| | - Mohammed Ali Gafar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
- Department of Pharmaceutics, Faculty of Pharmacy, University of Khartoum, Khartoum 11111, Sudan
| | - Lucy W Kiruri
- Department of Chemistry, Kenyatta University, P.O. Box 43844, Nairobi 00100, Kenya
| | - Usri H Ibrahim
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4300, South Africa
| | - Eman A Ismail
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| | - Nikita Devnarain
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag Durban X54001, South Africa
| |
Collapse
|
13
|
Amenta A, Comi S, Kravicz M, Sesana S, Antoniou A, Passarella D, Seneci P, Pellegrino S, Re F. A novel, glutathione-activated prodrug of pimasertib loaded in liposomes for targeted cancer therapy. RSC Med Chem 2024; 16:d4md00517a. [PMID: 39430954 PMCID: PMC11485093 DOI: 10.1039/d4md00517a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024] Open
Abstract
Pimasertib, a potent antiproliferative drug, has been extensively studied for treating cancers characterized by dysregulation in the ERK/MAPK signaling pathway, such as melanoma. However, its therapeutic efficacy would greatly benefit from an increased selectivity for tumour cells and a longer half-life. Such improvements may be achieved by combining the rational design of a prodrug with its encapsulation in a potential nanodelivery system. For this reason, we synthesized a glutathione (GSH)-responsive putative prodrug of pimasertib (PROPIMA), which contains a redox-sensitive disulphide linker that can be processed by GSH to activate pimasertib. The synthesis of PROPIMA and its in vitro biological activity on a human melanoma cell line as a model are described. The results showed that PROPIMA, either free or embedded in liposomes, selectively inhibits cell proliferation and cell viability, reducing by about 5-fold the levels of pERK. Additionally, PROPIMA shows stronger inhibition of the cancer cell migration than the parent drug.
Collapse
Affiliation(s)
- Arianna Amenta
- Department of Chemistry, University of Milan Milan Italy
| | - Susanna Comi
- School of Medicine and Surgery, University of Milano-Bicocca Monza Italy
| | - Marcelo Kravicz
- School of Medicine and Surgery, University of Milano-Bicocca Monza Italy
| | - Silvia Sesana
- School of Medicine and Surgery, University of Milano-Bicocca Monza Italy
| | - Antonia Antoniou
- Department of Pharmaceutical Sciences, University of Milan Milan Italy
| | | | | | - Sara Pellegrino
- Department of Pharmaceutical Sciences, University of Milan Milan Italy
| | - Francesca Re
- School of Medicine and Surgery, University of Milano-Bicocca Monza Italy
| |
Collapse
|
14
|
Verma VS, Pandey A, Jha AK, Badwaik HKR, Alexander A, Ajazuddin. Polyethylene Glycol-Based Polymer-Drug Conjugates: Novel Design and Synthesis Strategies for Enhanced Therapeutic Efficacy and Targeted Drug Delivery. Appl Biochem Biotechnol 2024; 196:7325-7361. [PMID: 38519751 DOI: 10.1007/s12010-024-04895-6] [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] [Accepted: 03/04/2024] [Indexed: 03/25/2024]
Abstract
Due to their potential to enhance therapeutic results and enable targeted drug administration, polymer-drug conjugates that use polyethylene glycol (PEG) as both the polymer and the linker for drug conjugation have attracted much research. This study seeks to investigate recent developments in the design and synthesis of PEG-based polymer-drug conjugates, emphasizing fresh ideas that fill in existing knowledge gaps and satisfy the increasing need for more potent drug delivery methods. Through an extensive review of the existing literature, this study identifies key challenges and proposes innovative strategies for future investigations. The paper presents a comprehensive framework for designing and synthesizing PEG-based polymer-drug conjugates, including rational molecular design, linker selection, conjugation methods, and characterization techniques. To further emphasize the importance and adaptability of PEG-based polymer-drug conjugates, prospective applications are highlighted, including cancer treatment, infectious disorders, and chronic ailments.
Collapse
Affiliation(s)
- Vinay Sagar Verma
- Faculty of Pharmaceutical Sciences, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai, Durg, Chhattisgarh, 490023, India
| | - Aakansha Pandey
- Faculty of Pharmaceutical Sciences, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India
| | - Arvind Kumar Jha
- Shri Shankaracharya Professional University, Junwani, Bhilai, 490020, Chhattisgarh, India
| | - Hemant Kumar Ramchandra Badwaik
- Shri Shankaracharya College of Pharmaceutical Sciences, Junwani, Bhilai, 490020, Chhattisgarh, India.
- Shri Shankaracharya Institute of Pharmaceutical Sciences and Research, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India.
| | - Amit Alexander
- Department of Pharmaceuticals, National Institute of Pharmaceutical Education and Research, Ministry of Chemical and Fertilizers, Guwahati, 781101, Assam, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai, Durg, Chhattisgarh, 490023, India.
| |
Collapse
|
15
|
Fahmy SA, Sedky NK, Hassan HAFM, Abdel-Kader NM, Mahdy NK, Amin MU, Preis E, Bakowsky U. Synergistic Enhancement of Carboplatin Efficacy through pH-Sensitive Nanoparticles Formulated Using Naturally Derived Boswellia Extract for Colorectal Cancer Therapy. Pharmaceutics 2024; 16:1282. [PMID: 39458611 PMCID: PMC11510476 DOI: 10.3390/pharmaceutics16101282] [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: 08/30/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Carboplatin (Cp) is a potent chemotherapeutic agent, but its effectiveness is constrained by its associated side effects. Frankincense, an oleo-gum resin from the Boswellia sacra tree, has demonstrated cytotoxic activity against cancer cells. This study explored the synergistic potential of nanoparticles formulated from Boswellia sacra methanolic extract (BME), to enhance the therapeutic efficacy of Cp at reduced doses. Nanoparticles were prepared via the nanoprecipitation method, loaded with Cp, and coated with positively charged chitosan (CS) for enhanced cell interaction, yielding Cp@CS/BME NPs with an average size of 160.2 ± 4.6 nm and a zeta potential of 12.7 ± 1.5 mV. In vitro release studies revealed a pH-sensitive release profile, with higher release rates at pH 5.4 than at pH 7.4, highlighting the potential for targeted drug delivery in acidic tumor environments. In vitro studies on HT-29 and Caco-2 colorectal cancer cell lines demonstrated the nanoformulation's ability to significantly increase Cp uptake and cytotoxic activity. Apoptosis assays further confirmed increased induction of cell death with Cp@CS/BME NPs. Cell-cycle analysis revealed that treatment with Cp@CS/BME NPs led to a significant increase in the sub-G1 phase, indicative of enhanced apoptosis, and a marked decrease in the G1-phase population coupled with an increased G2/M-phase arrest in both cell lines. Further gene expression analysis demonstrated a substantial downregulation of the anti-apoptotic gene Bcl-2 and an upregulation of the pro-apoptotic genes Bax, PUMA, and BID following treatment with Cp@CS/BME NPs. Thus, this study presents a promising and innovative strategy for enhancing the therapeutic efficacy of chemotherapeutic agents using naturally derived ingredients while limiting the side effects.
Collapse
Affiliation(s)
- Sherif Ashraf Fahmy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, R5 New Garden City, New Administrative Capital, Cairo 11835, Egypt
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany; (M.U.A.); (E.P.)
| | - Nada K. Sedky
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, R5 New Garden City, New Administrative Capital, Cairo 11835, Egypt; (N.K.S.); (N.M.A.-K.)
| | - Hatem A. F. M. Hassan
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, Kent ME4 4TB, UK
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt;
| | - Nour M. Abdel-Kader
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, R5 New Garden City, New Administrative Capital, Cairo 11835, Egypt; (N.K.S.); (N.M.A.-K.)
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo 11566, Egypt
| | - Noha Khalil Mahdy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt;
| | - Muhammad Umair Amin
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany; (M.U.A.); (E.P.)
| | - Eduard Preis
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany; (M.U.A.); (E.P.)
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany; (M.U.A.); (E.P.)
| |
Collapse
|
16
|
Mujahid K, Rana I, Suliman IH, Li Z, Wu J, He H, Nam J. Biomaterial-Based Sustained-Release Drug Formulations for Localized Cancer Immunotherapy. ACS APPLIED BIO MATERIALS 2024; 7:4944-4961. [PMID: 38050811 DOI: 10.1021/acsabm.3c00808] [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: 12/07/2023]
Abstract
Cancer immunotherapy has revolutionized clinical cancer treatments by taking advantage of the immune system to selectively and effectively target and kill cancer cells. However, clinical cancer immunotherapy treatments often have limited efficacy and/or present severe adverse effects associated primarily with their systemic administration. Localized immunotherapy has emerged to overcome these limitations by directly targeting accessible tumors via local administration, reducing potential systemic drug distribution that hampers drug efficacy and safety. Sustained-release formulations can prolong drug activity at target sites, which maximizes the benefits of localized immunotherapy to increase the therapeutic window using smaller dosages than those used for systemic injection, avoiding complications of frequent dosing. The performance of sustained-release formulations for localized cancer immunotherapy has been validated preclinically using various implantable and injectable scaffold platforms. This review introduces the sustained-release formulations developed for localized cancer immunotherapy and highlights their biomaterial-based platforms for representative classes, including inorganic scaffolds, natural hydrogels, synthetic hydrogels, and microneedle patches. The design rationale and other considerations are summarized for further development of biomaterials for the construction of optimal sustained-release formulations.
Collapse
Affiliation(s)
- Khizra Mujahid
- College of Pharmacy, Chonnam National University, Gwangju 61186, South Korea
| | - Isra Rana
- College of Pharmacy, Chonnam National University, Gwangju 61186, South Korea
| | | | - Zhen Li
- College of Pharmacy, Chonnam National University, Gwangju 61186, South Korea
| | - Jiang Wu
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Huacheng He
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, P. R. China
| | - Jutaek Nam
- College of Pharmacy, Chonnam National University, Gwangju 61186, South Korea
| |
Collapse
|
17
|
Raddi S, El Karmy B, Martinache O, Richert R, Colnot C, Grosgogeat B. Development of Chlorhexidine-loaded Lipid Nanoparticles Incorporated in a Bioceramic Endodontic Sealer. J Endod 2024; 50:1134-1142. [PMID: 38710385 DOI: 10.1016/j.joen.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 05/08/2024]
Abstract
INTRODUCTION This study aimed to assess BioRoot RCS (BR) incorporating liposomal chlorhexidine digluconate (CHX) for its antibacterial activity, drug release capacity, and physicochemical properties. METHODS Drug release of CHX liposomal formulations in combination with BR was evaluated spectrophotometrically and through mathematical release models for 30 days. A selected combination was evaluated for antimicrobial properties against Enterococcus faecalis biofilm growth on human dentin. Cytotoxicity was assessed following the ISO 10993-5:2019 standard on days 1, 3, and 7. Physicochemical properties were evaluated through setting time, Fourier transform infrared spectroscopy, solubility, contact angle, and film thickness. RESULTS From BR, liposomal CHX released up to 7-fold higher CHX than CHX solution (P < .05), following a triphasic drug release pattern compared to the CHX solution, which followed a quasi-Fickian diffusion. BR combined with a selected liposomal CHX completely inhibited E. faecalis biofilm growth compared to the combination of BR with CHX solution and the control group (P < .05). Liposomal CHX decreased the contact angle (P < .05) and solubility but increased cytotoxicity (P < .05) of BR, staying above the ISO threshold. None of the other physicochemical characteristics tested differed from BR (P > .05). CONCLUSION This liposomal formulation improved CHX release from BR, enhancing the antibacterial effectiveness. It presents a promising approach for local antibiofilm therapy in endodontics without substantially altering the physicochemical characteristics of BR.
Collapse
Affiliation(s)
- Samir Raddi
- CEMENTIC, Paris, France; Laboratoire des Multimatériaux et Interfaces, Université Claude Bernard Lyon 1, Villeurbanne, France.
| | | | | | - Raphael Richert
- Faculté d'Odontologie, Université Claude Bernard Lyon 1, Lyon, France; Hospices Civils de Lyon, Service de Consultations et de Traitements Dentaires, Unité de recherche clinique, Lyon, Lyon, France; University of Lyon, INSA-Lyon, CNRS UMR5259, LaMCoS, Villeurbanne, France
| | | | - Brigitte Grosgogeat
- Laboratoire des Multimatériaux et Interfaces, Université Claude Bernard Lyon 1, Villeurbanne, France; Faculté d'Odontologie, Université Claude Bernard Lyon 1, Lyon, France; Hospices Civils de Lyon, Service de Consultations et de Traitements Dentaires, Unité de recherche clinique, Lyon, Lyon, France
| |
Collapse
|
18
|
Kononenko V, Joukhan A, Bele T, Križaj I, Kralj S, Turk T, Drobne D. Gelatin nanoparticles loaded with 3-alkylpyridinium salt APS7, an analog of marine toxin, are a promising support in human lung cancer therapy. Biomed Pharmacother 2024; 177:117007. [PMID: 38906020 DOI: 10.1016/j.biopha.2024.117007] [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: 04/19/2024] [Revised: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024] Open
Abstract
This study demonstrates the potential of gelatin nanoparticles as a nanodelivery system for antagonists of nicotinic acetylcholine receptors (nAChRs) to improve chemotherapy efficacy and reduce off-target effects. Too often, chemotherapy for lung cancer does not lead to satisfactory results. Therefore, new approaches directed at multiple pharmacological targets in cancer therapy are being developed. Following the activation of nAChRs (e.g. by nicotine), cancer cells begin to proliferate and become more resistant to chemotherapy-induced apoptosis. This work shows that the 3-alkylpyridinium salt, APS7, a synthetic analog of a toxin from the marine sponge Haliclona (Rhizoneira) sarai, acts as an nAChR antagonist that inhibits the pro-proliferative and anti-apoptotic effects of nicotine on A549 human lung adenocarcinoma cells. In this study, gelatin-based nanoparticles filled with APS7 (APS7-GNPs) were prepared and their effects on A549 cells were compared with that of free APS7. Both APS7 and APS7-GNPs inhibited Ca2+ influx and increased the efficacy of cisplatin chemotherapy in nicotine-stimulated A549 cells. However, significant benefits from APS7-GNPs were observed - a stronger reduction in the proliferation of A549 lung cancer cells and a much higher selectivity in cytotoxicity towards cancer cells compared with non-tumorigenic lung epithelial BEAS-2B cells.
Collapse
Affiliation(s)
- Veno Kononenko
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana SI-1000, Slovenia.
| | - Ahmad Joukhan
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana SI-1000, Slovenia; Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana SI-1000, Slovenia
| | - Tadeja Bele
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, Ljubljana SI-1000, Slovenia; Faculty of medicine, University of Ljubljana, Vrazov trg 2, Ljubljana SI-1000, Slovenia
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, Ljubljana SI-1000, Slovenia
| | - Slavko Kralj
- Department for Materials Synthesis, Jožef Stefan Institute, Jamova 39, Ljubljana SI-1000, Slovenia
| | - Tom Turk
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana SI-1000, Slovenia
| | - Damjana Drobne
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana SI-1000, Slovenia.
| |
Collapse
|
19
|
Eluu SC, Obayemi JD, Yiporo D, Salifu AA, Oko AO, Onwudiwe K, Aina T, Oparah JC, Ezeala CC, Etinosa PO, Osafo SA, Ugwu MC, Esimone CO, Soboyejo WO. Luteinizing Hormone-Releasing Hormone (LHRH)-Conjugated Cancer Drug Delivery from Magnetite Nanoparticle-Modified Microporous Poly-Di-Methyl-Siloxane (PDMS) Systems for the Targeted Treatment of Triple Negative Breast Cancer Cells. J Funct Biomater 2024; 15:209. [PMID: 39194647 DOI: 10.3390/jfb15080209] [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: 06/24/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
This study presents LHRH conjugated drug delivery via a magnetite nanoparticle-modified microporous Poly-Di-Methyl-Siloxane (PDMS) system for the targeted suppression of triple-negative breast cancer cells. First, the MNP-modified PDMS devices are fabricated before loading with targeted and untargeted cancer drugs. The release kinetics from the devices are then studied before fitting the results to the Korsmeyer-Peppas model. Cell viability and cytotoxicity assessments are then presented using results from the Alamar blue assay. Apoptosis induction is then elucidated using flow cytometry. The in vitro drug release studies demonstrated a sustained and controlled release of unconjugated drugs (Prodigiosin and paclitaxel) and conjugated drugs [LHRH conjugated paclitaxel (PTX+LHRH) and LHRH-conjugated prodigiosin (PG+LHRH)] from the magnetite nanoparticle modified microporous PDMS devices for 30 days at 37 °C, 41 °C, and 44 °C. At 24, 48, 72, and 96 h, the groups loaded with conjugated drugs (PG+LHRH and PTX+LHRH) had a significantly higher (p < 0.05) percentage cell growth inhibition than the groups loaded with unconjugated drugs (PG and PTX). Additionally, throughout the study, the MNP+PDMS (without drug) group exhibited a steady rise in the percentage of cell growth inhibition. The flow cytometry results revealed a high incidence of early and late-stage apoptosis. The implications of the results are discussed for the development of biomedical devices for the localized and targeted release of cancer drugs that can prevent cancer recurrence following tumor resection.
Collapse
Affiliation(s)
- Stanley C Eluu
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Ifite Awka 420110, Nigeria
- Department of Biotechnology, Ebonyi State University, Abakaliki 480101, Nigeria
| | - John D Obayemi
- Department of Mechanical and Material Science Engineering, Higgins Lab, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA 01609, USA
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Gateway Park Life Sciences and Bioengineering Centre, 60 Prescott Street, Worcester, MA 01609, USA
| | - Danyuo Yiporo
- Department of Mechanical Engineering, Ashesi University, Berekuso PMB CT3, Ghana
- Department of Mechanical Engineering, Academic City University College, Haatso-Accra P.O. Box AD 421, Ghana
| | - Ali A Salifu
- Department of Engineering, Morrissey College of Arts and Science, Boston College, Chestnut Hill, MA 02467, USA
| | - Augustine O Oko
- Department of Biotechnology, Ebonyi State University, Abakaliki 480101, Nigeria
- Department of Biology and Biotechnology, David Umahi Federal University of Health Sciences, Uburu 480101, Nigeria
| | - Killian Onwudiwe
- Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Toyin Aina
- Department of Biomedical Engineering, Collage of Engineering, Afe Babalola University, KM 8.5 Afe Babalola Way, Ado-Ekiti 360001, Nigeria
| | - Josephine C Oparah
- Department of Material Science, African University of Science and Technology, Km 10 Airport Road, Abuja 900107, Nigeria
| | - Chukwudi C Ezeala
- Department of Material Science, African University of Science and Technology, Km 10 Airport Road, Abuja 900107, Nigeria
- Department of Biotechnology, Worcester State University, Worcester, MA 01602, USA
| | - Precious O Etinosa
- Department of Mechanical and Material Science Engineering, Higgins Lab, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA 01609, USA
| | - Sarah A Osafo
- Department of Material Science and Engineering, University of Ghana, Legon, Accra P.O. Box LG 1181, Ghana
- Biomaterial Science Department, Dental School, College of Health Sciences, University of Ghana, Korle bu, Accra P.O. Box KB 52, Ghana
| | - Malachy C Ugwu
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Ifite Awka 420110, Nigeria
| | - Charles O Esimone
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Ifite Awka 420110, Nigeria
| | - Winston O Soboyejo
- Department of Mechanical and Material Science Engineering, Higgins Lab, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA 01609, USA
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Gateway Park Life Sciences and Bioengineering Centre, 60 Prescott Street, Worcester, MA 01609, USA
- Department of Engineering, SUNY Polytechnic Institute,100 Seymour Rd, Utica, NY 13502, USA
| |
Collapse
|
20
|
Lee JS, Oh E, Oh H, Kim S, Ok S, Sa J, Lee JH, Shin YC, Bae YS, Choi CY, Lee S, Kwon HK, Yang S, Choi WI. Tacrolimus-loaded chitosan-based nanoparticles as an efficient topical therapeutic for the effective treatment of atopic dermatitis symptoms. Int J Biol Macromol 2024; 273:133005. [PMID: 38866268 DOI: 10.1016/j.ijbiomac.2024.133005] [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: 08/22/2023] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Atopic dermatitis (AD) is a chronic cutaneous disease with a complex underlying mechanism, and it cannot be completely cured. Thus, most treatment strategies for AD aim at relieving the symptoms. Although corticosteroids are topically applied to alleviate AD, adverse side effects frequently lead to the withdrawal of AD therapy. Tacrolimus (TAC), a calcineurin inhibitor, has been used to treat AD, but its high molecular weight and insolubility in water hinder its skin permeability. Herein, we developed and optimized TAC-loaded chitosan-based nanoparticles (TAC@CNPs) to improve the skin permeability of TAC by breaking the tight junctions in the skin. The prepared nanoparticles were highly loadable and efficient and exhibited appropriate characteristics for percutaneous drug delivery. TAC@CNP was stable for 4 weeks under physiological conditions. CNP released TAC in a controlled manner, with enhanced skin penetration observed. In vitro experiments showed that CNP was non-toxic to keratinocyte (HaCaT) cells, and TAC@CNP dispersed in an aqueous solution was as anti-proliferative as TAC solubilized in a good organic solvent. Importantly, an in vivo AD mouse model revealed that topical TAC@CNP containing ~1/10 of the dose of TAC found in commercially used Protopic® Ointment exhibited similar anti-inflammatory activity to that of the commercial product. TAC@CNP represents a potential therapeutic strategy for the management of AD.
Collapse
Affiliation(s)
- Jin Sil Lee
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea; School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Cheomdan-gwagiro, 123, Buk-gu, Gwangju 61005, Republic of Korea
| | - Eunjeong Oh
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| | - Hyeryeon Oh
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea; School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Cheomdan-gwagiro, 123, Buk-gu, Gwangju 61005, Republic of Korea
| | - Sunghyun Kim
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Subin Ok
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Junseo Sa
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | | | - Yong Chul Shin
- SKINMED Co Ltd., Daejeon 34028, Republic of Korea; Amicogen Inc, 64 Dongburo 1259, Jinsung, Jinju 52621, Republic of Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Cheol Yong Choi
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Siyoung Yang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Won Il Choi
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| |
Collapse
|
21
|
Mahboubi Kancha M, Mehrabi M, Aghaie F, Bitaraf FS, Dehghani F, Bernkop-Schnürch A. Preparation and characterization of PVA/chitosan nanofibers loaded with Dragon's blood or poly helixan as wound dressings. Int J Biol Macromol 2024; 272:132844. [PMID: 38834119 DOI: 10.1016/j.ijbiomac.2024.132844] [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: 11/25/2023] [Revised: 05/22/2024] [Accepted: 05/31/2024] [Indexed: 06/06/2024]
Abstract
Nanofibers have been investigated in regenerative medicine. Dragon's blood (DB)- and poly helixan PF (PHPF) are natural materials used in cosmetics. Herein, we generated DB- and PHPF-loaded polyvinyl alcohol/chitosan (PVA/CS/DB and PVA/CS/PHPF, respectively) nanofibers. PVA/CS/DB and PVA/CS/PHPF nanofibers had an average diameter of 547.5 ± 17.13 and 521 ± 24.67 nm, respectively as assessed by SEM, and a degradation rate of 43.1 and 47.6 % after 14 days, respectively. PVA/CS/DB and PVA/CS/PHPF nanofibers had a hemolysis rate of 0.10 and 0.39 %, respectively, and a water vapor transmission rate of ∼2200 g.m-2.day-1. These nanofibers exhibited favorable antimicrobial activity against Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, and Bacillus subtilis in vitro. PVA/CS/DB and PVA/CS/PHPF nanofibers demonstrated a sustained release of 77.91 and 76.55 % over 72 h. PVA/CS/DB and PVA/CS/PHPF nanofibers had a high rate of cytocompatibility and significantly improved the viability of NIH/3T3 cells as compared with free drugs or unloaded nanofibers. Histological inspection via H&E and Verhoeff's staining demonstrated PVA/CS/DB and PVA/CS/PHPF nanofibers enhanced the wound healing and damaged tissue recovery of unsplinted wound models by promoting epithelial layer formation, collagen deposition, and enhancing the presence of fibroblasts. Conclusively, PVA/CS/DB and PVA/CS/PHPF can be introduced as potential wound dressing candidates with favorable properties.
Collapse
Affiliation(s)
- Maral Mahboubi Kancha
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran; Department of Medical Nanotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohsen Mehrabi
- Department of Medical Nanotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Faeze Aghaie
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran; Department of Medical Biotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Fatemeh Sadat Bitaraf
- Department of Medical Biotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Farzaneh Dehghani
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, Innsbruck 6020, Austria
| |
Collapse
|
22
|
Alhudaithi SS, Abul Kalam M, Binobaid L, Ali R, Almutairi MM, Qamar W, Bin Hithlayn H, Almutairi A, Alshememry AK. Sorafenib and Piperine co-loaded PLGA nanoparticles: Development, characterization, and anti-cancer activity against hepatocellular carcinoma cell line. Saudi Pharm J 2024; 32:102064. [PMID: 38633710 PMCID: PMC11022100 DOI: 10.1016/j.jsps.2024.102064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/06/2024] [Indexed: 04/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) exhibits high mortality rates in the advanced stage (>90 %). Sorafenib (SORA) is a targeted therapy approved for the treatment of advanced HCC; however, the reported response rate to such a therapeutic is suboptimal (<3%). Piperine (PIP) is an alkaloid demonstrated to exert a direct tumoricidal activity in HCC and improve the pharmacokinetic profiles of anticancer drugs including SORA. In this study, we developed a strategy to improve efficacy outcomes in HCC using PIP as an add-on treatment to support the first-line therapy SORA using biodegradable Poly (D, L-Lactide-co-glycolide, PLGA) nanoparticles (NPs). SORA and PIP (both exhibit low aqueous solubility) were co-loaded into PLGA NPs (PNPs) and stabilized with various concentrations of polyvinyl alcohol (PVA). The SORA and PIP-loaded PNPs (SP-PNPs) were characterized using Fourier Transform Infrared (FTIR) Spectroscopy, X-ray Powder Diffraction (XRD), Dynamic Light Scattering (DLS), and Scanning Electron Microscopy (SEM), Release of these drugs from SP-PNPs was investigated in vitro at both physiological and acidic pH, and kinetic models were employed to assess the mechanism of drug release. The in vitro efficacy of SP-PNPs against HCC cells (HepG2) was also evaluated. FTIR and XRD analyses revealed that the drugs encapsulated in PNPs were in an amorphous state, with no observed chemical interactions among the drugs or excipients. Assessment of drug release in vitro at pH 5 and 7.4 showed that SORA and PIP loaded in PNPs with 0.5 % PVA were released in a sustained manner, unlike pure drugs, which exhibited relatively fast release. SP-PNPs with 0.5 % PVA were spherical, had an average size of 224 nm, and had a high encapsulation efficiency (SORA ∼ 82 %, PIP ∼ 79 %), as well as superior cytotoxicity compared to SORA monotherapy in vitro. These results suggest that combining PIP with SORA using PNPs may be an effective strategy for the treatment of HCC and may set the stage for a comprehensive in vivo study to evaluate the efficacy and safety of this novel formulation using a murine HCC model.
Collapse
Affiliation(s)
- Sulaiman S. Alhudaithi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohd Abul Kalam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Lama Binobaid
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Raisuddin Ali
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohammed M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Wajhul Qamar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Hessa Bin Hithlayn
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Atheer Almutairi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdullah K. Alshememry
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
23
|
Kimura T, Okada K, Morohashi Y, Kato Y, Mori M, Kato H, Matsumoto T, Shimoyama S. Quantification of Unencapsulated Drug in Target Tissues Demonstrates Pharmacological Properties and Therapeutic Effects of Liposomal Topotecan (FF-10850). Pharm Res 2024; 41:795-806. [PMID: 38536615 PMCID: PMC11024016 DOI: 10.1007/s11095-023-03652-4] [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: 08/09/2023] [Accepted: 12/27/2023] [Indexed: 04/18/2024]
Abstract
PURPOSE Quantifying unencapsulated drug concentrations in tissues is crucial for understanding the mechanisms underlying the efficacy and safety of liposomal drugs; however, the methodology for this has not been fully established. Herein, we aimed to investigate the enhanced therapeutic potential of a pegylated liposomal formulation of topotecan (FF-10850) by analyzing the concentrations of the unencapsulated drug in target tissues, to guide the improvement of its dosing regimen. METHODS We developed a method for measuring unencapsulated topotecan concentrations in tumor and bone marrow interstitial fluid (BM-ISF) and applied this method to pharmacokinetic assessments. The ratios of the area under the concentration-time curves (AUCs) between tumor and BM-ISF were calculated for total and unencapsulated topotecan. DNA damage and antitumor effects of FF-10850 or non-liposomal topotecan (TPT) were evaluated in an ES-2 mice xenograft model. RESULTS FF-10850 exhibited a much larger AUC ratio between tumor and BM-ISF for unencapsulated topotecan (2.96), but not for total topotecan (0.752), than TPT (0.833). FF-10850 promoted milder DNA damage in the bone marrow than TPT; however, FF-10850 and TPT elicited comparable DNA damage in the tumor. These findings highlight the greater tumor exposure to unencapsulated topotecan and lower bone marrow exposure to FF-10850 than TPT. The dosing regimen was successfully improved based on the kinetics of unencapsulated topotecan and DNA damage. CONCLUSIONS Tissue pharmacokinetics of unencapsulated topotecan elucidated the favorable pharmacological properties of FF-10850. Evaluation of tissue exposure to an unencapsulated drug with appropriate pharmacodynamic markers can be valuable in optimizing liposomal drugs and dosing regimens.
Collapse
Affiliation(s)
- Toshifumi Kimura
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan.
| | - Ken Okada
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Yasushi Morohashi
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Mikinaga Mori
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Hiroshi Kato
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Takeshi Matsumoto
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-Machi, Ashigarakami-Gun, Kanagawa, 258-8577, Japan
| | - Susumu Shimoyama
- FUJIFILM Pharmaceuticals U.S.A., Inc, One Broadway, Cambridge, MA, 02142, USA
| |
Collapse
|
24
|
Sakkal M, Arafat M, Yuvaraju P, Beiram R, AbuRuz S. Preparation and Characterization of Theophylline Controlled Release Matrix System Incorporating Poloxamer 407, Stearyl Alcohol, and Hydroxypropyl Methylcellulose: A Novel Formulation and Development Study. Polymers (Basel) 2024; 16:643. [PMID: 38475326 DOI: 10.3390/polym16050643] [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: 01/24/2024] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Theophylline (THN), a bronchodilator with potential applications in emerging conditions like COVID-19, requires a controlled-release delivery system due to its narrow therapeutic range and short half-life. This need is particularly crucial as some existing formulations demonstrate impaired functionality. This study aims to develop a new 12-h controlled-release matrix system (CRMS) in the form of a capsule to optimize dosing intervals. METHODS CRMSs were developed using varying proportions of poloxamer 407 (P-407), stearyl alcohol (STA), and hydroxypropyl methylcellulose (HPMC) through the fusion technique. Their in vitro dissolution profiles were then compared with an FDA-approved THN drug across different pH media. The candidate formulation underwent characterization using X-ray diffraction, scanning electron microscopy, Fourier transform infrared spectroscopy, differential scanning calorimetry, and thermogravimetric analysis. Additionally, a comprehensive stability study was conducted. RESULTS In vitro studies showed that adjusting the concentrations of excipients effectively controlled drug release. Notably, the CRMS formulation 15 (CRMS-F15), which was composed of 30% P-407, 30% STA, and 10% HPMC, closely matched the 12 h controlled-release profile of an FDA-approved drug across various pH media. Characterization techniques verified the successful dispersion of the drug within the matrix. Furthermore, CRMS-F15 maintained a consistent controlled drug release and demonstrated stability under a range of storage conditions. CONCLUSIONS The newly developed CRMS-F15 achieved a 12 h controlled release, comparable to its FDA-approved counterpart.
Collapse
Affiliation(s)
- Molham Sakkal
- College of Pharmacy, Al Ain University, Al Ain P.O. Box 64141, United Arab Emirates
| | - Mosab Arafat
- College of Pharmacy, Al Ain University, Al Ain P.O. Box 64141, United Arab Emirates
| | - Priya Yuvaraju
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Rami Beiram
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Salahdein AbuRuz
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
25
|
Dartora VFC, Passos JS, Costa-Lotufo LV, Lopes LB, Panitch A. Thermosensitive Polymeric Nanoparticles for Drug Co-Encapsulation and Breast Cancer Treatment. Pharmaceutics 2024; 16:231. [PMID: 38399285 PMCID: PMC10892816 DOI: 10.3390/pharmaceutics16020231] [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: 01/14/2024] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Despite advances in breast cancer treatment, there remains a need for local management of noninvasive, low-grade ductal carcinoma in situ (DCIS). These focal lesions are well suited for local intraductal treatment. Intraductal administration supported target site drug retention, improved efficacy, and reduced systemic exposure. Here, we used a poly(N-isopropyl acrylamide, pNIPAM) nanoparticle delivery system loaded with cytotoxic piplartine and an MAPKAP Kinase 2 inhibitor (YARA) for this purpose. For tumor environment targeting, a collagen-binding peptide SILY (RRANAALKAGELYKSILYGSG-hydrazide) was attached to pNIPAM nanoparticles, and the nanoparticle diameter, zeta potential, drug loading, and release were assessed. The system was evaluated for cytotoxicity in a 2D cell culture and 3D spheroids. In vivo efficacy was evaluated using a chemical carcinogenesis model in female Sprague-Dawley rats. Nanoparticle delivery significantly reduced the IC50 of piplartine (4.9 times) compared to the drug in solution. The combination of piplartine and YARA in nanoparticles further reduced the piplartine IC50 (~15 times). Treatment with these nanoparticles decreased the in vivo tumor incidence (5.2 times). Notably, the concentration of piplartine in mammary glands treated with nanoparticles (35.3 ± 22.4 μg/mL) was substantially higher than in plasma (0.7 ± 0.05 μg/mL), demonstrating targeted drug retention. These results indicate that our nanocarrier system effectively reduced tumor development with low systemic exposure.
Collapse
Affiliation(s)
- Vanessa Franco Carvalho Dartora
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-900, Brazil; (V.F.C.D.); (J.S.P.); (L.V.C.-L.); (L.B.L.)
- Department of Biomedical Engineering, College of Engineering, University of California Davis, Davis, CA 95616, USA
| | - Julia S. Passos
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-900, Brazil; (V.F.C.D.); (J.S.P.); (L.V.C.-L.); (L.B.L.)
| | - Leticia V. Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-900, Brazil; (V.F.C.D.); (J.S.P.); (L.V.C.-L.); (L.B.L.)
| | - Luciana B. Lopes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-900, Brazil; (V.F.C.D.); (J.S.P.); (L.V.C.-L.); (L.B.L.)
| | - Alyssa Panitch
- Department of Biomedical Engineering, College of Engineering, University of California Davis, Davis, CA 95616, USA
- Wallace H. Coulter Department of Biomedical Engineering, College of Engineering, Georgia Institute of Technology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
26
|
Sa P, Mohapatra P, Swain SS, Khuntia A, Sahoo SK. Phytochemical-Based Nanomedicine for Targeting Tumor Microenvironment and Inhibiting Cancer Chemoresistance: Recent Advances and Pharmacological Insights. Mol Pharm 2023; 20:5254-5277. [PMID: 37596986 DOI: 10.1021/acs.molpharmaceut.3c00286] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2023]
Abstract
Cancer remains the leading cause of death and rapidly evolving disease worldwide. The understanding of disease pathophysiology has improved through advanced research investigation, and several therapeutic strategies are being used for better cancer treatment. However, the increase in cancer relapse and metastatic-related deaths indicate that available therapies and clinically approved chemotherapy drugs are not sufficient to combat cancer. Further, the constant crosstalk between tumor cells and the tumor microenvironment (TME) is crucial for the development, progression, metastasis, and therapeutic response to tumors. In this regard, phytochemicals with multimodal targeting abilities can be used as an alternative to current cancer therapy by inhibiting cancer survival pathways or modulating TME. However, due to their poor pharmacokinetics and low bioavailability, the success of phytochemicals in clinical trials is limited. Therefore, developing phytochemical-based nanomedicine or phytonanomedicine can improve the pharmacokinetic profile of these phytochemicals. Herein, the molecular characteristics and pharmacological insights of the proposed phytonanomedicine in cancer therapy targeting tumor tissue and altering the characteristics of cancer stem cells, chemoresistance, TME, and cancer immunity are well discussed. Further, we have highlighted the clinical perspective and challenges of phytonanomedicine in filling the gap in potential cancer therapeutics using various nanoplatforms. Overall, we have discussed how clinical success and pharmacological insights could make it more beneficial to boost the concept of nanomedicine in the academic and pharmaceutical fields to counter cancer metastases and drug resistance.
Collapse
Affiliation(s)
- Pratikshya Sa
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, NCR Delhi, India
| | - Priyanka Mohapatra
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, NCR Delhi, India
| | | | - Auromira Khuntia
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, NCR Delhi, India
| | | |
Collapse
|
27
|
Madkhali OA. Drug Delivery of Gelatin Nanoparticles as a Biodegradable Polymer for the Treatment of Infectious Diseases: Perspectives and Challenges. Polymers (Basel) 2023; 15:4327. [PMID: 37960007 PMCID: PMC10648051 DOI: 10.3390/polym15214327] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
In recent years, there has been a growing interest in the use of gelatin nanoparticles (GNPs) for the treatment of infectious diseases. The inherent properties of these nanoparticles make them attractive options for drug delivery. Their biocompatibility ensures that they can interact with biological systems without causing adverse reactions, while their biodegradability ensures that they can break down harmlessly in the body once their function is performed. Furthermore, their capacity for controlled drug release ensures that therapeutic agents can be delivered over a sustained period, thereby enhancing treatment efficacy. This review examines the current landscape of GNP-based drug delivery, with a specific focus on its potential applications and challenges in the context of infectious diseases. Key challenges include controlling drug release rates, ensuring nanoparticle stability under physiological conditions, scaling up production while maintaining quality, mitigating potential immunogenic reactions, optimizing drug loading efficiency, and tracking the biodistribution and clearance of GNPs in the body. Despite these hurdles, GNPs hold promising potential in the realm of infectious disease treatment. Ongoing research and innovation are essential to overcome these obstacles and completely harness the potential of GNPs in clinical applications.
Collapse
Affiliation(s)
- Osama A Madkhali
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45124, Saudi Arabia
| |
Collapse
|
28
|
Bai X, Tang S, Butterworth S, Tirella A. Design of PLGA nanoparticles for sustained release of hydroxyl-FK866 by microfluidics. BIOMATERIALS ADVANCES 2023; 154:213649. [PMID: 37820459 DOI: 10.1016/j.bioadv.2023.213649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 09/25/2023] [Accepted: 09/30/2023] [Indexed: 10/13/2023]
Abstract
The use of nanoparticle (NP) delivery systems in cancer treatment has received significant interest, however use of such systems in delivery of cytotoxic chemotherapy agents can be limited by low encapsulation efficiency and burst release of the cytotoxin, as well issues with throughput and reproducibility during the fabrication of drug-loaded NPs. In this study, we used a hydrodynamic flow-focusing microfluidic system to successfully produce poly(lactic-co-glycolic acid) (PLGA) NPs. The physico-chemical properties of PLGA NPs were controlled by changing the manufacturing parameters, such as flow rate ratio, total flow rate, PLGA and surfactant concentration. The NAMPT inhibitor-polymer conjugate, hydroxyl-FK866-PLGA, was synthesized and used to fabricate hydroxyl-FK866-PLGA NPs for the formulation of localized delivery systems able to release low doses of cytotoxins and enhance the efficacy of NAMPT inhibitors. Hydroxyl-FK866-PLGA NPs were prepared with optimized fabrication parameters, having average Z-size of 128 ± 8 nm (PDI < 0.2), ζ-potential of -14.8 ± 5.3 mV and high encapsulation efficiency (98.6 ± 5.8 %). The pH-dependent release of hydroxyl-FK866 was monitored over time in conditions mimicking the normal (pH 7.4) and inflamed/tumor (pH 6.4) microenvironments, observing a sustained release pattern (over two months) without any initial burst release. Finally, toxicity of hydroxyl-FK866-PLGA NPs were tested in selected human cell lines, the human leukemia monocytic cell line (THP-1), and the human triple negative breast cancer cell line (MDA-MB-231). Our work suggests that microfluidic systems are a promising technology for a rapid and efficient manufacturing of PLGA-based NPs for the controlled release of cytotoxins. Moreover, the use of drug-polymer conjugates is an effective approach for the manufacturing of polymeric NPs enabling high encapsulation efficiency and a prolonged and sustained pH-dependent drug release.
Collapse
Affiliation(s)
- Xue Bai
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Siyuan Tang
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Sam Butterworth
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Annalisa Tirella
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK; BIOtech Center for Biomedical Technologies, Department of Industrial Engineering, University of Trento, Via delle Regole 101, 38123 Trento, Italy.
| |
Collapse
|
29
|
Moya-Garcia CR, Li-Jessen NYK, Tabrizian M. Chitosomes Loaded with Docetaxel as a Promising Drug Delivery System to Laryngeal Cancer Cells: An In Vitro Cytotoxic Study. Int J Mol Sci 2023; 24:9902. [PMID: 37373051 DOI: 10.3390/ijms24129902] [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: 04/29/2023] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Current delivery of chemotherapy, either intra-venous or intra-arterial, remains suboptimal for patients with head and neck tumors. The free form of chemotherapy drugs, such as docetaxel, has non-specific tissue targeting and poor solubility in blood that deters treatment efficacy. Upon reaching the tumors, these drugs can also be easily washed away by the interstitial fluids. Liposomes have been used as nanocarriers to enhance docetaxel bioavailability. However, they are affected by potential interstitial dislodging due to insufficient intratumoral permeability and retention capabilities. Here, we developed and characterized docetaxel-loaded anionic nanoliposomes coated with a layer of mucoadhesive chitosan (chitosomes) for the application of chemotherapy drug delivery. The anionic liposomes were 99.4 ± 1.5 nm in diameter with a zeta potential of -26 ± 2.0 mV. The chitosan coating increased the liposome size to 120 ± 2.2 nm and the surface charge to 24.8 ± 2.6 mV. Chitosome formation was confirmed via FTIR spectroscopy and mucoadhesive analysis with anionic mucin dispersions. Blank liposomes and chitosomes showed no cytotoxic effect on human laryngeal stromal and cancer cells. Chitosomes were also internalized into the cytoplasm of human laryngeal cancer cells, indicating effective nanocarrier delivery. A higher cytotoxicity (p < 0.05) of docetaxel-loaded chitosomes towards human laryngeal cancer cells was observed compared to human stromal cells and control treatments. No hemolytic effect was observed on human red blood cells after a 3 h exposure, proving the proposed intra-arterial administration. Our in vitro results supported the potential of docetaxel-loaded chitosomes for locoregional chemotherapy delivery to laryngeal cancer cells.
Collapse
Affiliation(s)
- Christian R Moya-Garcia
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, 3775 Rue University, Montreal, QC H3A 2B4, Canada
| | - Nicole Y K Li-Jessen
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, 3775 Rue University, Montreal, QC H3A 2B4, Canada
- School of Communication Sciences and Disorders, McGill University, 2001 Av. McGill College #8, Montréal, QC H3A 1G1, Canada
- Department of Otolaryngology-Head and Neck Surgery, McGill University Health Centre, 1001 Decarie Blvd., Montreal, QC H4A 3J1, Canada
- Research Institute of the McGill University Health Centre, 1001 Decarie Blvd., Montreal, QC H4A 3J1, Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, 3775 Rue University, Montreal, QC H3A 2B4, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, 2001 Av. McGill College, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
30
|
Vogelaar A, Marcotte S, Cheng J, Oluoch B, Zaro J. Use of Microfluidics to Prepare Lipid-Based Nanocarriers. Pharmaceutics 2023; 15:pharmaceutics15041053. [PMID: 37111539 PMCID: PMC10144662 DOI: 10.3390/pharmaceutics15041053] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Lipid-based nanoparticles (LBNPs) are an important tool for the delivery of a diverse set of drug cargoes, including small molecules, oligonucleotides, and proteins and peptides. Despite their development over the past several decades, this technology is still hindered by issues with the manufacturing processes leading to high polydispersity, batch-to-batch and operator-dependent variability, and limits to the production volumes. To overcome these issues, the use of microfluidic techniques in the production of LBNPs has sharply increased over the past two years. Microfluidics overcomes many of the pitfalls seen with conventional production methods, leading to reproducible LBNPs at lower costs and higher yields. In this review, the use of microfluidics in the preparation of various types of LBNPs, including liposomes, lipid nanoparticles, and solid lipid nanoparticles for the delivery of small molecules, oligonucleotides, and peptide/protein drugs is summarized. Various microfluidic parameters, as well as their effects on the physicochemical properties of LBNPs, are also discussed.
Collapse
Affiliation(s)
- Alicia Vogelaar
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Samantha Marcotte
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Jiaqi Cheng
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Benazir Oluoch
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Jennica Zaro
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
31
|
Salama ABM, Salem YY, Mohamed TMA. Controlled and Targeted Drug Delivery Using Smart Nanovectors. INTERNATIONAL JOURNAL OF DRUG DISCOVERY AND PHARMACOLOGY 2023; 2:84-90. [PMID: 39524321 PMCID: PMC11545783 DOI: 10.53941/ijddp.0201010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The conventional drug delivery systems have several limitations, such as the high frequency of administration, several off-target effects, and the need for tissue specificity. Recently, smart drug shuttles have emerged, and the nano applications provided a new opportunity for advancing the drug delivery system to become tissue targeted and decrease the frequency of administration. The recent development of nanovectors as drug carriers has gone through several steps of evolution that ended with the development of logic-embedded nanovectors. Here, we summarize the different types of nanovectors and their applications in various clinical situations, and finally, we spot the light on the future of this area of research.
Collapse
Affiliation(s)
- Abou Bakr M. Salama
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY 40208, U.S.A
- Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Yasmin Y. Salem
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY 40208, U.S.A
- Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Tamer M. A. Mohamed
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY 40208, U.S.A
- Envirome Institute, Centre for Cardiometabolic Sciences, Department of Medicine, University of Louisville, KY 40208, U.S.A
- Department of Bioengineering, Speed School of Engineering, University of Louisville, KY 40208, U.S.A
- Department of Pharmacology and Toxicology, University of Louisville, KY 40208, U.S.A
- Institute of Cardiovascular Sciences, University of Manchester M13 9PL, U.K
| |
Collapse
|
32
|
Hyaluronic Acid-Coated Chitosan Nanoparticles as an Active Targeted Carrier of Alpha Mangostin for Breast Cancer Cells. Polymers (Basel) 2023; 15:polym15041025. [PMID: 36850308 PMCID: PMC9965946 DOI: 10.3390/polym15041025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Alpha mangostin (AM) has potential anticancer properties for breast cancer. This study aims to assess the potential of chitosan nanoparticles coated with hyaluronic acid for the targeted delivery of AM (AM-CS/HA) against MCF-7 breast cancer cells. AM-CS/HA showed a spherical shape with an average diameter of 304 nm, a polydispersity index of 0.3, and a negative charge of 24.43 mV. High encapsulation efficiency (90%) and drug loading (8.5%) were achieved. AM released from AM-CS/HA at an acidic pH of 5.5 was higher than the physiological pH of 7.4 and showed sustained release. The cytotoxic effect of AM-CS/HA (IC50 4.37 µg/mL) on MCF-7 was significantly higher than AM nanoparticles without HA coating (AM-CS) (IC50 4.48 µg/mL) and AM (IC50 5.27 µg/mL). These findings suggest that AM-CS/HA enhances AM cytotoxicity and has potential applications for breast cancer therapy.
Collapse
|
33
|
Bibliometric mapping of solid lipid nanoparticles research (2012–2022) using VOSviewer. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2023. [DOI: 10.1016/j.medntd.2023.100217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023] Open
|
34
|
Jampilek J, Kralova K. Anticancer Applications of Essential Oils Formulated into Lipid-Based Delivery Nanosystems. Pharmaceutics 2022; 14:2681. [PMID: 36559176 PMCID: PMC9781429 DOI: 10.3390/pharmaceutics14122681] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
The use of natural compounds is becoming increasingly popular among patients, and there is a renewed interest among scientists in nature-based bioactive agents. Traditionally, herbal drugs can be taken directly in the form of teas/decoctions/infusions or as standardized extracts. However, the disadvantages of natural compounds, especially essential oils, are their instability, limited bioavailability, volatility, and often irritant/allergenic potential. However, these active substances can be stabilized by encapsulation and administered in the form of nanoparticles. This brief overview summarizes the latest results of the application of nanoemulsions, liposomes, solid lipid nanoparticles, and nanostructured lipid carriers used as drug delivery systems of herbal essential oils or used directly for their individual secondary metabolites applicable in cancer therapy. Although the discussed bioactive agents are not typical compounds used as anticancer agents, after inclusion into the aforesaid formulations improving their stability and bioavailability and/or therapeutic profile, they indicated anti-tumor activity and became interesting agents with cancer treatment potential. In addition, co-encapsulation of essential oils with synthetic anticancer drugs into nanoformulations with the aim to achieve synergistic effect in chemotherapy is discussed.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
- Department of Chemical Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 783 71 Olomouc, Czech Republic
| | - Katarina Kralova
- Institute of Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
| |
Collapse
|