251
|
Baruah N, Halder P, Koley H, Katti DS. Stable Recombinant Invasion Plasmid Antigen C (IpaC)-Based Single Dose Nanovaccine for Shigellosis. Mol Pharm 2022; 19:3884-3893. [PMID: 36122190 DOI: 10.1021/acs.molpharmaceut.2c00378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Shigellosis, caused by the bacteria Shigella, is the leading cause of bacterial diarrhea and the second leading cause of diarrheal death among children under the age of five. Unfortunately, Shigella strains have acquired resistance to antibiotics, and a commercial vaccine is yet to be available. We have previously demonstrated that Shigella dysenteriae serotype 1 (Sd1)-based recombinant, stabilized, "invasion plasmid antigen C" (IpaC; 42 kDa) protein can induce robust immune responses in BALB/c mice against a challenge of a high dose of heterologous Shigella when immunized via three intranasal doses of IpaC without an adjuvant. In this work, in order to reduce the frequency of dosing and increase possible patient compliance, based on our previous screening, the minimum protective dose of stabilized IpaC (20 μg) was encapsulated in biodegradable polymeric poly(lactide-co-glycolide) nanoparticles (∼370 nm) and intranasally administered in BALB/c mice in a single dose. Interestingly, a single intranasal dose of the developed vaccine particles encapsulating only 20 μg of Sd1 IpaC led to a temporal increase in the antibody production with an improved cytokine response compared to free IpaC administered three times as described in our previous report. Upon intraperitoneal challenge with a high dose of heterologous Shigella flexneri 2a (common in circulation), the immunized animals were protected from diarrhea, lethargy, and weight loss with ∼67% survival, while all the control animals died by 36 h of the challenge. Overall, the developed nanovaccine could be explored as a potential noninvasive, cross-protective, single-dose, single-antigen Shigella vaccine amenable for scale-up and eventual mass immunization.
Collapse
Affiliation(s)
- Namrata Baruah
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India.,The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera & Enteric Diseases, Kolkata, West Bengal 700010, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera & Enteric Diseases, Kolkata, West Bengal 700010, India
| | - Dhirendra S Katti
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India.,The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
252
|
Varga N, Seres L, Kovács NA, Turcsányi Á, Juhász Á, Csapó E. Serum albumin/hyaluronic acid nanoconjugate: Evaluation of concentration-dependent structural changes to form an efficient drug carrier particle. Int J Biol Macromol 2022; 220:1523-1531. [PMID: 36122775 DOI: 10.1016/j.ijbiomac.2022.09.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Norbert Varga
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary
| | - László Seres
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary
| | - Nikolett Alexandra Kovács
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary
| | - Árpád Turcsányi
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary
| | - Ádám Juhász
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary
| | - Edit Csapó
- MTA-SZTE Lendület "Momentum" Noble Metal Nanostructures Research Group, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary; Interdisciplinary Excellence Center, Department of Physical Chemistry and Materials Science, University of Szeged, H-6720 Rerrich B. sqr. 1, Szeged, Hungary.
| |
Collapse
|
253
|
Kaur G, Kumar P. Ibuprofen tagged Imine RT-COF1 as customisable vehicle for controlled drug delivery application. INORG CHEM COMMUN 2022. [DOI: 10.1016/j.inoche.2022.110043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
254
|
Zhang F, Zhang C, Fu S, Liu H, Han M, Fan X, Zhang H, Li W. Amphiphilic Cationic Peptide-Coated PHA Nanosphere as an Efficient Vector for Multiple-Drug Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3024. [PMID: 36080060 PMCID: PMC9457696 DOI: 10.3390/nano12173024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/08/2022] [Accepted: 08/26/2022] [Indexed: 06/15/2023]
Abstract
Amphiphilic core-shell (ACS) nanoparticles are gaining increasing research interest for multi-drug delivery in cancer therapy. In this work, a new cationic peptide-coated PHA nanosphere was prepared by self-assembly of a hydrophobic core of biodegradable poly (3-hydroxybutyrate-co-3-hydroxyhexanoate) (PHBHHx) and a hydrophilic shell of fusion proteins of PHA granule-associated protein (PhaP) and cationic peptide RALA through a strong hydrophobic effect. The hydrophobic drug curcumin (Cur) was encapsulated in PHBHHx nanoparticles. The chemotherapy drug 5-fluorouracil (5-FU) was administered in the form of its metabolite oligomeric 5-fluorodeoxyuridine (FUdR). Fifteen consecutive FUdR (FUdR15S) were adsorbed on the surface of PHBHHx nanoparticles by electrostatic interaction with RALA to form Cur@PHBX-PR/FUdR15S. Such amphiphilic cationic nanospheres had 88.3% EE of Cur and the drug loading of Cur and FUdR were 7.8% and 12.1%. The dual-drug-loaded nanospheres showed a time-differential release of Cur and FUdR. In addition, Cur@PHBX-PR/FUdR15S exhibited excellent anticancer activity and played a vital role in promoting the synergistic effect of FUdR and Cur in gastric cancer cells. The exploration of antitumor mechanisms demonstrated that Cur improved the activity of apoptosis-related proteins and cancer cells sensitized to FUdR. This amphiphilic core-shell system can serve as a general platform for sequential delivery of multiple drugs to treat several cancer cells.
Collapse
Affiliation(s)
- Fanghua Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
- Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, School of Science, Tianjin University, Tianjin 300072, China
| | - Chao Zhang
- Department of Life Science, Hengshui University, Hengshui 053000, China
| | - Shuangqing Fu
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Huandi Liu
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Mengnan Han
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Xueyu Fan
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Honglei Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Wei Li
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| |
Collapse
|
255
|
Lee DN, Kim YR, Kim Y, Park BJ, Lee SJ, Kim SJ, Shin JH. Therapeutic Potency of NO Loaded into Anticancer Copper Metal-Organic Framework through Nonclassical Hydrogen Bonding. ACS APPLIED BIO MATERIALS 2022; 5:4301-4309. [PMID: 36041482 DOI: 10.1021/acsabm.2c00501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Metal-organic frameworks (MOFs) are potential exogenous scaffolds for therapeutic nitric oxide (NO) delivery because they can store drug or bioactive gas molecules within pores or on active metal sites. Herein, we employed a Cu-MOF coordinated with glutarate (glu) and 1,2-bis(4-pyridyl)ethane (bpa) to obtain NO-loaded Cu-MOF (NO⊂Cu-MOF). NO loading transformed the space group of Cu-MOF from monoclinic C2/c to triclinic P-1 through nonclassical hydrogen bonding with glu and bpa. Cu-MOF showed good stability in deionized water and phosphate-buffered saline. NO⊂Cu-MOF released up to 1.10 μmol mg-1 NO over 14.6 h at 37 °C, which is suitable for therapeutic applications. NO⊂Cu-MOF showed moderate biocompatibility with L-929 cells and significant anticancer activity against HeLa cells, suggesting an apoptosis-mediated cell death mechanism. These insights into NO bonding modes with Cu-MOF that enable controlled NO release can inspire the design of functional MOFs as hybrid NO donors for drug delivery.
Collapse
Affiliation(s)
- Do Nam Lee
- Ingenium College of Liberal Arts (Chemistry), Kwangwoon University, Seoul01897, Republic of Korea
| | - Yeong Rim Kim
- Department of Chemistry, Kwangwoon University, Seoul01897, Republic of Korea
| | - Youngmee Kim
- NanoBio-Energy Materials Center and Department of Chemistry and Nano Science, Ewha Womans University, Seoul03760, Republic of Korea
| | - Bong Joo Park
- Department of Electrical and Biological Physics, Kwangwoon University, Seoul01897, Republic of Korea
| | - Su Jung Lee
- Ingenium College of Liberal Arts (Chemistry), Kwangwoon University, Seoul01897, Republic of Korea
| | - Sung-Jin Kim
- NanoBio-Energy Materials Center and Department of Chemistry and Nano Science, Ewha Womans University, Seoul03760, Republic of Korea
| | - Jae Ho Shin
- Department of Chemistry, Kwangwoon University, Seoul01897, Republic of Korea
| |
Collapse
|
256
|
Tewari AK, Upadhyay SC, Kumar M, Pathak K, Kaushik D, Verma R, Bhatt S, Massoud EES, Rahman MH, Cavalu S. Insights on Development Aspects of Polymeric Nanocarriers: The Translation from Bench to Clinic. Polymers (Basel) 2022; 14:3545. [PMID: 36080620 PMCID: PMC9459741 DOI: 10.3390/polym14173545] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 02/06/2023] Open
Abstract
Scientists are focusing immense attention on polymeric nanocarriers as a prominent delivery vehicle for several biomedical applications including diagnosis of diseases, delivery of therapeutic agents, peptides, proteins, genes, siRNA, and vaccines due to their exciting physicochemical characteristics which circumvent degradation of unstable drugs, reduce toxic side effects through controlled release, and improve bioavailability. Polymers-based nanocarriers offer numerous benefits for in vivo drug delivery such as biocompatibility, biodegradability, non-immunogenicity, active drug targeting via surface modification, and controlled release due to their pH-and thermosensitive characteristics. Despite their potential for medicinal use, regulatory approval has been achieved for just a few. In this review, we discuss the historical development of polymers starting from their initial design to their evolution as nanocarriers for therapeutic delivery of drugs, peptides, and genes. The review article also expresses the applications of polymeric nanocarriers in the pharmaceutical and medical industry with a special emphasis on oral, ocular, parenteral, and topical application of drugs, peptides, and genes over the last two decades. The review further examines the practical, regulatory, and clinical considerations of the polymeric nanocarriers, their safety issues, and directinos for future research.
Collapse
Affiliation(s)
- Akhilesh Kumar Tewari
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana, India
| | - Satish Chandra Upadhyay
- Formulation Research and Development, Mankind Research Centre, Manesar, Gurugram 122050, Haryana, India
| | - Manish Kumar
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana, India
| | - Kamla Pathak
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah 206130, Uttar Pradesh, India
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, Haryana, India
| | - Ravinder Verma
- Department of Pharmacy, G.D. Goenka University, Sohna Road, Gurugram 122103, Haryana, India
| | - Shailendra Bhatt
- Department of Pharmacy, G.D. Goenka University, Sohna Road, Gurugram 122103, Haryana, India
| | - Ehab El Sayed Massoud
- Biology Department, Faculty of Science and Arts in Dahran Aljnoub, King Khalid University, Abha 62529, Saudi Arabia
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 61413, Saudi Arabia
- Agriculture Research Centre, Soil, Water and Environment Research Institute, Giza 3725004, Egypt
| | - Md. Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| |
Collapse
|
257
|
Marei HE. Multimodal targeting of glioma with functionalized nanoparticles. Cancer Cell Int 2022; 22:265. [PMID: 35999629 PMCID: PMC9396820 DOI: 10.1186/s12935-022-02687-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/16/2022] [Indexed: 11/10/2022] Open
Abstract
The most common and aggressive primitive intracranial tumor of the central nervous system is the glioma. The blood–brain barrier (BBB) has proven to be a significant obstacle to the effective treatment of glioma. To effectively treat glioma, different ways have been used to cross the BBB to deliver drugs to the brain. Drug delivery through nanocarriers proves to be an effective and non-invasive technique for the treatment of glioma and has great potential in the treatment of glioma. In this review, we will provide an overview of nanocarrier-mediated drug delivery and related glioma therapy. Nanocarrier-mediated drug delivery techniques to cross the BBB (liposomes, micelles, inorganic systems, polymeric nanoparticles, nanogel system, and biomimetic nanoparticles) are explored. Finally, the use of nanotherapeutic approaches in the treatment of glioblastoma including chemotherapy, radiotherapy, photothermal therapy, gene therapy, glioma genome editing, immunotherapy, chimeric antigen receptor (CAR) T-cells, immune checkpoint modulators, immune photothermal therapy, vaccine-based immunotherapy, and combination therapy is summarized. Furthermore, this article offers various views on the clinical applicability of nanomedicine.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| |
Collapse
|
258
|
Ajalli N, Pourmadadi M, Yazdian F, Rashedi H, Navaei-Nigjeh M, Díez-Pascual AM. Chitosan/Gamma-Alumina/Fe3O4@5-FU Nanostructures as Promising Nanocarriers: Physiochemical Characterization and Toxicity Activity. Molecules 2022; 27:molecules27175369. [PMID: 36080138 PMCID: PMC9458215 DOI: 10.3390/molecules27175369] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 01/07/2023] Open
Abstract
Today, cancer treatment is an important issue in the medical world due to the challenges and side effects of ongoing treatment procedures. Current methods can be replaced with targeted nano-drug delivery systems to overcome such side effects. In the present work, an intelligent nano-system consisting of Chitosan (Ch)/Gamma alumina (γAl)/Fe3O4 and 5-Fluorouracil (5-FU) was synthesized and designed for the first time in order to influence the Michigan Cancer Foundation-7 (MCF-7) cell line in the treatment of breast cancer. Physico-chemical characterization of the nanocarriers was carried out using X-ray diffraction (XRD), Fourier-transform infrared spectroscopy (FTIR), vibrating sample magnetometry (VSM), dynamic light scattering (DLS), and scanning electron microscopy (SEM). SEM analysis revealed smooth and homogeneous spherical nanoparticles. The high stability of the nanoparticles and their narrow size distribution was confirmed by DLS. The results of the loading study demonstrated that these nano-systems cause controlled, stable, and pH-sensitive release in cancerous environments with an inactive targeting mechanism. Finally, the results of MTT and flow cytometry tests indicated that this nano-system increased the rate of apoptosis induction on cancerous masses and could be an effective alternative to current treatments.
Collapse
Affiliation(s)
- Narges Ajalli
- Department of Chemical Engineering, Faculty of Engineering, University of Tehran, Tehran 1417935840, Iran
| | - Mehrab Pourmadadi
- Department of Chemical Engineering, Faculty of Engineering, University of Tehran, Tehran 1417935840, Iran
| | - Fatemeh Yazdian
- Department of Life Science Engineering, Faculty of New Science and Technologies, University of Tehran, Tehran 1439956191, Iran
- Correspondence: (F.Y.); (H.R.); (A.M.D.-P.)
| | - Hamid Rashedi
- Department of Chemical Engineering, Faculty of Engineering, University of Tehran, Tehran 1417935840, Iran
- Correspondence: (F.Y.); (H.R.); (A.M.D.-P.)
| | - Mona Navaei-Nigjeh
- Pharmaceutical Sciences Research Center, Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417613151, Iran
- Department of Pharmaceutical Biomaterials, Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Ana M. Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805 Alcalá de Henares, Madrid, Spain
- Correspondence: (F.Y.); (H.R.); (A.M.D.-P.)
| |
Collapse
|
259
|
Eudragit® L100/Polyvinyl Alcohol Nanoparticles Impregnated Mucoadhesive Films as Ocular Inserts for Controlled Delivery of Erythromycin: Development, Characterization and In Vivo Evaluation. Biomedicines 2022; 10:biomedicines10081917. [PMID: 36009463 PMCID: PMC9405482 DOI: 10.3390/biomedicines10081917] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 11/26/2022] Open
Abstract
The fast elimination of drugs from the cornea is one of many challenges associated with the topical administration of conventional dosage forms. The present manuscript aimed to prepare modified-release inserts containing erythromycin (ERY) to enhance drug delivery and address the aforementioned limitation. Film formulations were developed using Eudragit® L100 (EUD) and Polyvinyl Alcohol (PVA) polymers. ERY-loaded EUD-based nanoparticles were developed by the colloidal dispersion method using PVA as the emulsifier. The film-casting method was applied to form the mucoadhesive films using sodium alginate, gelatin, cyclodextrin-α, and β as polymeric film matrices. Different physicochemical properties of the optimized formulations and in vitro release profiles were evaluated. The in vivo evaluation was performed by collecting tear samples of rabbits using a novel, non-invasive method following the administration of inserts in the cul-de-sac. The ERY amount was assayed using a microbiological assay. The developed films showed prolonged in vitro and in vivo release profiles over five to six days; they had suitable physicochemical properties and a tensile strength of 2–3 MPa. All formulations exhibited antibacterial efficacy against E. coli and S. aureus with more than 20 mm diameter of inhibited growth zones. None of the formulations caused irritation to the rabbit’s eye. The inserts showed promising pharmacokinetics with AUC0–120 of 30,000–36,000 µg·h/mL, a Cmax of more than 1800 µg/mL at 4 h, and maintained drug concentration over the threshold of 5 µg/mL during the following 120 h of study. Nanoparticle-containing, mucoadhesive films could be fabricated as ocular inserts and can prolong the topical ocular delivery of ERY.
Collapse
|
260
|
Amphotericin B-PEG Conjugates of ZnO Nanoparticles: Enhancement Antifungal Activity with Minimal Toxicity. Pharmaceutics 2022; 14:pharmaceutics14081646. [PMID: 36015271 PMCID: PMC9415822 DOI: 10.3390/pharmaceutics14081646] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/17/2022] Open
Abstract
Amphotericin B (AMB) is commonly used to treat life-threatening systemic fungal infections. AMB formulations that are more efficient and less nephrotoxic are currently unmet needs. In the current study, new ZnO-PEGylated AMB (ZnO-AMB-PEG) nanoparticles (NPs) were synthesized and their antifungal effects on the Candida spp. were investigated. The size and zeta potential values of AMB-PEG and ZnO-AMB-PEG NPs were 216.2 ± 26.9 to 662.3 ± 24.7 nm and −11.8 ± 2.02 to −14.2 ± 0.94 mV, respectively. The FTIR, XRD, and EDX spectra indicated that the PEG-enclosed AMB was capped by ZnO, and SEM images revealed the ZnO distribution on the surface NPs. In comparison to ZnO-AMB NPs and free AMB against C.albicans and C.neoformans, ZnO-AMB-PEG NPs significantly reduced the MIC and MFC. After a week of single and multiple dosage, the toxicity was investigated utilizing in vitro blood hemolysis, in vivo nephrotoxicity, and hepatic functions. ZnO-AMB-PEG significantly lowered WBC count and hematocrit concentrations when compared to AMB and ZnO-AMB. RBC count and hemoglobulin content, on the other hand, were unaltered. ZnO-AMB-PEG considerably lowered creatinine and blood urea nitrogen (BUN) levels when compared to AMB and ZnO-AMB. The difference in liver function indicators was determined to be minor by all formulae. These findings imply that ZnO-AMB-PEG could be utilized in the clinic with little nephrotoxicity, although more research is needed to determine the formulation’s in vivo efficacy.
Collapse
|
261
|
Aegerter N, Luijten A, Massella D, Ermanni P. Production of highly concentrated commodity thermoplastic NP suspensions with 3D printed confined impinging jet mixers and efficient downstream operations. POWDER TECHNOL 2022. [DOI: 10.1016/j.powtec.2022.117835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
262
|
Samy M, Abdallah HM, Awad HM, Ayoub MMH. In vitro release and cytotoxicity activity of 5-fluorouracil entrapped polycaprolactone nanoparticles. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-021-03804-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
263
|
|
264
|
Moore TL, Cook AB, Bellotti E, Palomba R, Manghnani P, Spanò R, Brahmachari S, Di Francesco M, Palange AL, Di Mascolo D, Decuzzi P. Shape-specific microfabricated particles for biomedical applications: a review. Drug Deliv Transl Res 2022; 12:2019-2037. [PMID: 35284984 PMCID: PMC9242933 DOI: 10.1007/s13346-022-01143-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2022] [Indexed: 12/13/2022]
Abstract
The storied history of controlled the release systems has evolved over time; from degradable drug-loaded sutures to monolithic zero-ordered release devices and nano-sized drug delivery formulations. Scientists have tuned the physico-chemical properties of these drug carriers to optimize their performance in biomedical/pharmaceutical applications. In particular, particle drug delivery systems at the micron size regime have been used since the 1980s. Recent advances in micro and nanofabrication techniques have enabled precise control of particle size and geometry-here we review the utility of microplates and discoidal polymeric particles for a range of pharmaceutical applications. Microplates are defined as micrometer scale polymeric local depot devices in cuboid form, while discoidal polymeric nanoconstructs are disk-shaped polymeric particles having a cross-sectional diameter in the micrometer range and a thickness in the hundreds of nanometer range. These versatile particles can be used to treat several pathologies such as cancer, inflammatory diseases and vascular diseases, by leveraging their size, shape, physical properties (e.g., stiffness), and component materials, to tune their functionality. This review highlights design and fabrication strategies for these particles, discusses their applications, and elaborates on emerging trends for their use in formulations.
Collapse
Affiliation(s)
- Thomas L Moore
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy.
| | - Alexander B Cook
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Elena Bellotti
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Roberto Palomba
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Purnima Manghnani
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Raffaele Spanò
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Sayanti Brahmachari
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Martina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Anna Lisa Palange
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Daniele Di Mascolo
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| |
Collapse
|
265
|
Dey SK, Pradhan A, Roy T, Das S, Chattopadhyay D, Maiti Choudhury S. Biogenic polymer-encapsulated diosgenin nanoparticles: Biodistribution, pharmacokinetics, cellular internalization, and anticancer potential in breast cancer cells and tumor xenograft. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
266
|
Tran Q, Pham TL, Shin HJ, Shin J, Shin N, Kwon HH, Park H, Kim SI, Choi SG, Wu J, Ngo VTH, Park JB, Kim DW. Targeting spinal microglia with fexofenadine-loaded nanoparticles prolongs pain relief in a rat model of neuropathic pain. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 44:102576. [PMID: 35714922 DOI: 10.1016/j.nano.2022.102576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
Targeting microglial activation is emerging as a clinically promising drug target for neuropathic pain treatment. Fexofenadine, a histamine receptor 1 antagonist, is a clinical drug for the management of allergic reactions as well as pain and inflammation. However, the effect of fexofenadine on microglial activation and pain behaviors remains elucidated. Here, we investigated nanomedicinal approach that targets more preferentially microglia and long-term analgesics. Fexofenadine significantly abolished histamine-induced microglial activation. The fexofenadine-encapsulated poly(lactic-co-glycolic acid) nanoparticles (Fexo NPs) injection reduced the pain sensitivity of spinal nerve ligation rats in a dose-dependent manner. This alleviation was sustained for 4 days, whereas the effective period by direct fexofenadine injection was 3 h. Moreover, Fexo NPs inhibited microglial activation, inflammatory signaling, cytokine release, and a macrophage phenotype shift towards the alternative activated state in the spinal cord. These results show that Fexo NPs exhibit drug repositioning promise as a long-term treatment modality for neuropathic pain.
Collapse
Affiliation(s)
- Quangdon Tran
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; Molecular Biology Laboratory, Department of Medical Laboratories, Hai Phong International Hospital, Hai Phong City #18000, Viet Nam
| | - Thuy Linh Pham
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; Department of Histology & Embryology, Hai Phong University of Medicine & Pharmacy, Hai Phong 042-12, Viet Nam
| | - Hyo Jung Shin
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Nara Shin
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Hyeok Hee Kwon
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Seoung Gyu Choi
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Junhua Wu
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Van T H Ngo
- Graduate Department of Healthcare Science, Dainam University, Viet Nam
| | - Jin Bong Park
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; Department of Physiology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea.
| |
Collapse
|
267
|
TLR2 agonistic lipopeptide enriched PLGA nanoparticles as combinatorial drug delivery vehicle. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
268
|
Ma J, Yu DH, Zhao D, Huang T, Dong M, Wang T, Yin HT. Poly-Lactide-Co-Glycolide-Polyethylene Glycol-Ginsenoside Rg3-Ag Exerts a Radio-Sensitization Effect in Non-Small Cell Lung Cancer. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Radiotherapy is an effective anti-cancer therapy for patients with non-small cell lung cancer (NSCLC), however, the prognosis is unsatisfactory owing to radio-resistance and toxicity. It is crucial to improve radiotherapy efficacy. Ag nanoparticles (NPs) and ginsenoside Rg3 (Rg3) exerted
antitumor and radio-sensitization effects. Therefore, we investigated whether poly-lactide-co-glycolide-polyethylene glycol (PLGA-PEG)-Rg3-Ag will function as a noninvasive, tracing, radiotherapy sensitizer. The morphology of NPs was visualized with transmission electron microscopy (TEM).
The drug loading content, encapsulation efficiency, and cumulative drug release of Rg3 was determined by HPLC. Cellular uptake of NPs in A549 and SPCA-1 was measured by immunostaining. The radio-sensitization effect of PLGA-PEG-Rg3-Ag in vitro was determined in A549 by detecting proliferation,
colony formation, and apoptosis with CCK-8, clonogenic survival assay, and flow cytometry, while in vivo was determined in nude mice by testing the body weight and tumor volume. PLGA-PEG-Rg3-Ag exerted radio-sensitization effect by reducing cell proliferation and colony formation while
enhancing cell apoptosis in A549; reduced tumor volume in nude mice. PLGA-PEG-Rg3-Ag exhibits radio-sensitization effects in NSCLC.
Collapse
Affiliation(s)
- Jun Ma
- Radiotherapy Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Da-Hai Yu
- Radiotherapy Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Di Zhao
- Radiotherapy Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Teng Huang
- Radiotherapy Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Min Dong
- Radiotherapy Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Ting Wang
- Radiotherapy Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Hai-Tao Yin
- Radiotherapy Department, Xuzhou Central Hospital, Xuzhou, 221000, Jiangsu Province, China
| |
Collapse
|
269
|
Liu L, Ma Q, Wang S, Gao Y, Zhu C, Zhao W, Sun W, Ma H, Sun Y. Efficient epidermal delivery of antibiotics by self-assembled lecithin/chitosan nanoparticles for enhanced therapy on epidermal bacterial infections. Int J Biol Macromol 2022; 218:568-579. [PMID: 35902014 DOI: 10.1016/j.ijbiomac.2022.07.165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022]
Abstract
The treatment for epidermal bacterial infections has become a primary healthy concern, producing a significant therapeutic challenge. Here we present a facile strategy to fabricate lecithin/chitosan nanoparticles (LCNPs) for efficient epidermal drug delivery over epidermal bacterial infections. The central rotatable composite design method was used for the optimization of the preparation, and that the optimal size (212.63 ± 1.95 nm) was obtained via analysis of variance (ANOVA). The prepared CIP-LCNPs show an average diameter of 325.9 ± 7.4 nm and a zeta potential of 26.6 ± 1.2 mV. Antibiotics can be well encapsulated in LCNPs and its release kinetics is studied with cumulative release of 93.81 ± 2.05 % for 48 h. The hemolytic activity, cytotoxicity, and skin irritation are further investigated. The zones of inhibition are 2.16 ± 0.04 cm and 2.92 ± 0.03 cm for Escherichia coli and Staphylococcus aureus, respectively. Moreover, in vitro permeation studies demonstrate that LCNPs can increase the accumulation of antibiotics in the epidermis with retention ratio 2-3 fold higher than commercial formulations. The in vivo result over epidermal-infected wound demonstrates the superior therapeutic effects of LCNPs. The developed LCNPs represent an important advance in fabricating therapeutic materials for enhanced therapy over epidermal bacterial infections.
Collapse
Affiliation(s)
- Lijun Liu
- School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Qingming Ma
- School of Pharmacy, Qingdao University, Qingdao 266071, China.
| | - Suning Wang
- Tongliao Market Detection and Testing Center, Tongliao 028000, China
| | - Yang Gao
- School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Chunrong Zhu
- School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Wenbin Zhao
- School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Wentao Sun
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China.
| | - Haifeng Ma
- Department of Geriatrics, Zibo Municipal Hospital, Zibo 255400, China.
| | - Yong Sun
- School of Pharmacy, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
270
|
Development of graphene oxide nanoscrolls imparted nano-delivery system for the sustained release of gallic acid. APPLIED NANOSCIENCE 2022. [DOI: 10.1007/s13204-022-02582-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
271
|
Roig-Soriano X, Souto EB, Elmsmari F, Garcia ML, Espina M, Duran-Sindreu F, Sánchez-López E, González Sánchez JA. Nanoparticles in Endodontics Disinfection: State of the Art. Pharmaceutics 2022; 14:1519. [PMID: 35890414 PMCID: PMC9316632 DOI: 10.3390/pharmaceutics14071519] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 01/27/2023] Open
Abstract
Endodontic-related diseases constitute the fourth most expensive pathologies in industrialized countries. Specifically, endodontics is the part of dentistry focused on treating disorders of the dental pulp and its consequences. In order to treat these problems, especially endodontic infections, dental barriers and complex root canal anatomy should be overcome. This constitutes an unmet medical need since the rate of successful disinfection with the currently marketed drugs is around 85%. Therefore, nanoparticles constitute a suitable alternative in order to deliver active compounds effectively to the target site, increasing their therapeutic efficacy. Therefore, in the present review, an overview of dental anatomy and the barriers that should be overcome for effective disinfection will be summarized. In addition, the versatility of nanoparticles for drug delivery and their specific uses in dentistry are comprehensively discussed. Finally, the latest findings, potential applications and state of the art nanoparticles with special emphasis on biodegradable nanoparticles used for endodontic disinfection are also reviewed.
Collapse
Affiliation(s)
- Xavier Roig-Soriano
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
- REQUIMTE/UCIBIO, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Firas Elmsmari
- Department of Clinical Sciences, College of Dentistry, Ajman University, University Street Al Jerf 1, Ajman 346, United Arab Emirates
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, University Street Al Jerf 1, Ajman 346, United Arab Emirates
| | - Maria Luisa Garcia
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
| | - Marta Espina
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
| | - Fernando Duran-Sindreu
- Department of Endodontics, Faculty of Dentistry, Universitat Internacional de Catalunya, 08017 Barcelona, Spain
| | - Elena Sánchez-López
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
- Unit of Synthesis and Biomedical Applications of Peptides, IQAC-CSIC, 08034 Barcelona, Spain
| | | |
Collapse
|
272
|
Caraway CA, Gaitsch H, Wicks EE, Kalluri A, Kunadi N, Tyler BM. Polymeric Nanoparticles in Brain Cancer Therapy: A Review of Current Approaches. Polymers (Basel) 2022; 14:2963. [PMID: 35890738 PMCID: PMC9322801 DOI: 10.3390/polym14142963] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022] Open
Abstract
Translation of novel therapies for brain cancer into clinical practice is of the utmost importance as primary brain tumors are responsible for more than 200,000 deaths worldwide each year. While many research efforts have been aimed at improving survival rates over the years, prognosis for patients with glioblastoma and other primary brain tumors remains poor. Safely delivering chemotherapeutic drugs and other anti-cancer compounds across the blood-brain barrier and directly to tumor cells is perhaps the greatest challenge in treating brain cancer. Polymeric nanoparticles (NPs) are powerful, highly tunable carrier systems that may be able to overcome those obstacles. Several studies have shown appropriately-constructed polymeric NPs cross the blood-brain barrier, increase drug bioavailability, reduce systemic toxicity, and selectively target central nervous system cancer cells. While no studies relating to their use in treating brain cancer are in clinical trials, there is mounting preclinical evidence that polymeric NPs could be beneficial for brain tumor therapy. This review includes a variety of polymeric NPs and how their associated composition, surface modifications, and method of delivery impact their capacity to improve brain tumor therapy.
Collapse
Affiliation(s)
- Chad A. Caraway
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Hallie Gaitsch
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
- NIH-Oxford-Cambridge Scholars Program, Wellcome—MRC Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK
| | - Elizabeth E. Wicks
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
- University of Mississippi School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Anita Kalluri
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Navya Kunadi
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Betty M. Tyler
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| |
Collapse
|
273
|
Sarkar S, Thapa R, Naushin F, Gupta S, Bhar B, De R, Bhattacharya J. Antibiotic-Loaded Smart Platelet: A Highly Effective Invisible Mode of Killing Both Antibiotic-Sensitive and -Resistant Bacteria. ACS OMEGA 2022; 7:24102-24110. [PMID: 35874209 PMCID: PMC9301723 DOI: 10.1021/acsomega.1c07249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Microbial pathogenesis is considered one of the most critical health challenges worldwide. Although several antibiotics have been procured and used, the microbes often manage to escape and become resistant to antibiotics. Thus, the discovery of new antibiotics and designing smart approaches toward their delivery are of great importance. In many cases, the delivery agents using foreign chemicals like lipids or polymers induce immunogenic responses of varying degrees and are limited to a shorter circulatory time and burst release. In the current work, we have designed a novel antibiotic delivery system where the antibiotic is encapsulated into a blood component-platelet. Platelets have been previously reported as efficient drug delivery vehicles for targeting cancer cells. On the other hand, during platelet-bacterial interaction, platelets can act as covercytes. Keeping this in mind, smart antibiotic-loaded platelets have been used for killing bacterial cells. The loading of the antibiotic was done using its typical nature of engulfing surrounding small molecules. The water-soluble antibiotics were loaded directly into the platelet, whereas the hydrophobic antibiotics were preloaded in polycaprolactone (FDA-approved polymer)-based nanovesicles to make them solubilized prior to loading inside the platelets. The antibiotic-loaded platelets (containing hydrophilic antibiotics or hydrophobic antibiotic -encapsulated polymer nanoparticles) were found to be stable when studied through platelet aggregometry. The carrier showed bactericidal effects at a significantly lower concentration at which the free antibiotic has negligible efficacy. This could be attributed to the molecular confinement of the antibiotics inside the platelets, therefore causing localization of the drug and leading to efficient activity against bacteria. Interestingly, the smart antibiotic-loaded platelets were capable of killing the resistant strains too at the same lower concentration regime. Therefore, the antibiotic-loaded platelet could emerge as a potential strategy for efficient delivery of antibiotics with a significant reduction of the dose required to achieve the intended antibacterial efficacy. Moreover, this antibiotic delivery method can be very useful to minimize immunogenic responses due to antibiotic administration and to avoid the development of drug resistance due to the invisible mode of delivery.
Collapse
Affiliation(s)
- Sounik Sarkar
- School
of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Roshni Thapa
- School
of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
- Special
Centre for Molecular Medicine, Jawaharlal
Nehru University, New Delhi 110067, India
| | - Farzana Naushin
- School
of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Saurabh Gupta
- School
of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Biswajit Bhar
- Institute
of Hematology & Transfusion Medicine, Medical College and Hospital, Kolkata 700073, India
| | - Rajib De
- Haematology
Department, NRS Medical College, Kolkata 700014, India
| | | |
Collapse
|
274
|
Local retention efficacy of steroid-loaded PLGA microspheres in epidural injection. Sci Rep 2022; 12:12244. [PMID: 35851101 PMCID: PMC9293917 DOI: 10.1038/s41598-022-16359-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 07/08/2022] [Indexed: 11/18/2022] Open
Abstract
Long-term effects of epidural steroid injections for pain management require novel drug formulations that increase tissue retention time. Present study aimed to investigate the local retention of steroid-loaded poly(lactic-co-glycolic acid) (PLGA) microspheres in epidural injection using a rabbit model. Twenty rabbits were randomly assigned to a PLGA group (n = 10) and a triamcinolone acetonide (TA) group (n = 10). Each animal was injected with either TA-loaded PLGA microspheres or conventional TA suspension into the lumbar epidural space. The lumbar segments were then harvested from the sacrificed rabbits on day 1, week 1, 2, and 4 after the injection. On day 1, the residual steroid concentration (RSC) was lower in the PLGA group than in the TA group (5.03 ppm vs. 13.01 ppm). However, after a week, more steroids remained in the PLGA group (3.29 ppm vs. 0.58 ppm). After 2 weeks, fewer steroids remained in the PLGA group than in the TA group, although both contained less than 10% of the initial retention dose. This study shows that steroid-loaded PLGA tended to have higher steroid retention in tissue than the steroid itself at the first week after epidural injection. However, most of the steroids disappeared after 2 weeks in both groups.
Collapse
|
275
|
Chopra H, Bibi S, Singh I, Kamal MA, Islam F, Alhumaydhi FA, Emran TB, Cavalu S. Nanomedicines in the Management of Alzheimer's Disease: Current View and Future Prospects. Front Aging Neurosci 2022; 14:879114. [PMID: 35875806 PMCID: PMC9304964 DOI: 10.3389/fnagi.2022.879114] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/17/2022] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a kind of dementia that creates serious challenges for sufferers' memory, thinking, and behavior. It commonly targeting the aging population and decay the brain cells, despite attempts have been performed to enhance AD diagnostic and therapeutic techniques. Hence, AD remains incurable owing to its complex and multifactorial consequences and still there is lack of appropriate diagnostics/therapeutics option for this severe brain disorder. Therefore, nanotechnology is currently bringing new tools and insights to improve the previous knowledge of AD and ultimately may provide a novel treatment option and a ray of hope to AD patients. Here in this review, we highlighted the nanotechnologies-based findings for AD, in both diagnostic and therapeutic aspects and explained how advances in the field of nanotechnology/nanomedicine could enhance patient prognosis and quality of life. It is highly expected these emerging technologies could bring a research-based revolution in the field of neurodegenerative disorders and may assist their clinical experiments and develop an efficacious drug for AD also. The main aim of review is to showcase readers the recent advances in nanotechnology-based approaches for treatment and diagnosing of AD.
Collapse
Affiliation(s)
- Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Shabana Bibi
- Department of Biosciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
- Yunnan Herbal Laboratory, College of Ecology and Environmental Sciences, Yunnan University, Kunming, China
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- Enzymoics, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
276
|
Salari N, Faraji F, Torghabeh FM, Faraji F, Mansouri K, Abam F, Shohaimi S, Akbari H, Mohammadi M. Polymer-based drug delivery systems for anticancer drugs: A systematic review. Cancer Treat Res Commun 2022; 32:100605. [PMID: 35816909 DOI: 10.1016/j.ctarc.2022.100605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/21/2022] [Accepted: 07/02/2022] [Indexed: 06/15/2023]
Abstract
Recent advances in nanotechnology sciences lead to the development of new treatment approaches for various diseases such as cancer. Nanotechnology advances can potentially minimize the side effects of drugs through the employment of effective and controlled drug delivery systems (DDSs). Polymers are optimal tools providing drug delivery mechanisms through the unique features of pharmacokinetics, circulation time, biocompatibility, and biodegradability. This systematic review aimed to evaluate polymer-based DDSs for anticancer drugs and their various therapeutic applications in cancer treatment. This study was conducted with no time limitation by November 2021. Related articles were collected through a deep search in English and Persian databases of SID, MagIran, Scopus, Web Of Science (WoS), PubMed, Science Direct, and Google Scholar. Keywords included drug delivery system, anticancer agent, polymeric nanostructure-based drug delivery, polymer-based drug delivery, and polymeric system. As the results showed, polymeric nanoparticles (PNPs) have influential roles in cancer treatment than conventional chemotherapy procedures. PNPs can reduce cytotoxicity following chemotherapy drug administration, improve the solubility characteristics of these therapeutic agents and inhibit the rate of tumor growth.
Collapse
Affiliation(s)
- Nader Salari
- Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Mansouri Torghabeh
- Department of Physiology Sciences, Medical School, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Faraji
- Student research committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Centre, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farzaneh Abam
- Medical Biology Research Centre, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shamarina Shohaimi
- Department of Biology, Faculty of Science, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - Hakimeh Akbari
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Masoud Mohammadi
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran.
| |
Collapse
|
277
|
Yang B, Gomes Dos Santos A, Puri S, Bak A, Zhou L. The industrial design, translation, and development strategies for long-acting peptide delivery. Expert Opin Drug Deliv 2022; 19:1233-1245. [PMID: 35787229 DOI: 10.1080/17425247.2022.2098276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Peptides are widely recognized as therapeutic agents in the treatment of a wide range of diseases, such as cancer, diabetes etc. However, their use has been limited by their short half-life, due to significant metabolism by exo- and endo-peptidases as well as their inherent poor physical and chemical stability. Research with the aim of improving their half-life in the body, and thus improving patient compliance (by decreasing the frequency of injections) has gained significant attention. AREAS COVERED This review outlines the current landscape and industrial approaches to achieve extended peptide exposure and reduce dosing frequency. Emphasis is placed on identifying challenges in drug product manufacturing and desirable critical quality attributes that are essential for activity and safety, providing insights into chemistry and design aspects impacting peptide release, and summarizing important considerations for CMC developability assessments of sustained release peptide drugs. EXPERT OPINION Bring the patient and disease perspective early into development. Substantial advances have been made in the field of sustained delivery of peptides despite their complexity. The article will also highlight considerations for early-stage product design and development, providing an industrial perspective on risk mitigation in developing sustained release peptide drug products.
Collapse
Affiliation(s)
- Bin Yang
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Ana Gomes Dos Santos
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Sanyogitta Puri
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Annette Bak
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, USA
| | - Liping Zhou
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, USA
| |
Collapse
|
278
|
Azagury A, Baptista C, Milovanovic K, Shin H, Morello P, Perez-Rogers J, Goldenshtein V, Nguyen T, Markel A, Rege S, Hojsak S, Perl A, Jones C, Fife M, Furtado S, Mathiowitz E. Biocoating-A Critical Step Governing the Oral Delivery of Polymeric Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107559. [PMID: 35606684 PMCID: PMC9250634 DOI: 10.1002/smll.202107559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/11/2022] [Indexed: 05/13/2023]
Abstract
Decades of research into the topic of oral nanoparticle (NP) delivery has still not provided a clear consensus regarding which properties produce an effective oral drug delivery system. The surface properties-charge and bioadhesiveness-as well as in vitro and in vivo correlation seem to generate the greatest number of disagreements within the field. Herein, a mechanism underlying the in vivo behavior of NPs is proposed, which bridges the gaps between these disagreements. The mechanism relies on the idea of biocoating-the coating of NPs with mucus-which alters their surface properties, and ultimately their systemic uptake. Utilizing this mechanism, several coated NPs are tested in vitro, ex vivo, and in vivo, and biocoating is found to affect NPs size, zeta-potential, mucosal diffusion coefficient, the extent of aggregation, and in vivo/in vitro/ex vivo correlation. Based on these results, low molecular weight polylactic acid exhibits a 21-fold increase in mucosal diffusion coefficient after precoating as compared to uncoated particles, as well as 20% less aggregation, and about 30% uptake to the blood in vivo. These discoveries suggest that biocoating reduces negative NP charge which results in an enhanced mucosal diffusion rate, increased gastrointestinal retention time, and high systemic uptake.
Collapse
Affiliation(s)
- Aharon Azagury
- Noninvasive Biomimetic Drug Delivery Systems Lab, The Department of Chemical Engineering, Ariel Center for Applied Cancer Research (ACACR), Ariel University, Ramat HaGolan St 65, Ari'el, 40700000, Israel
| | - Cameron Baptista
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Kosta Milovanovic
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Hyeseon Shin
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Peter Morello
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - James Perez-Rogers
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Victoria Goldenshtein
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Travis Nguyen
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Arianna Markel
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Soham Rege
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Stephanie Hojsak
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Alexander Perl
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Carder Jones
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Megan Fife
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Stacia Furtado
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| | - Edith Mathiowitz
- Department of Pathology and Laboratory Medicine, Center of Biomedical Engineering, Brown University, 171 Meeting Street, Box G-B3, Providence, RI, 02912, USA
| |
Collapse
|
279
|
Mondal S, Das S, Mahapatra PK, Saha KD. Morin encapsulated chitosan nanoparticles (MCNPs) ameliorate arsenic induced liver damage through improvement of the antioxidant system and prevention of apoptosis and inflammation in mice. NANOSCALE ADVANCES 2022; 4:2857-2872. [PMID: 36132010 PMCID: PMC9419452 DOI: 10.1039/d2na00167e] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/30/2022] [Indexed: 05/31/2023]
Abstract
Chronic exposure to arsenic over a period of time induces toxicity, primarily in the liver but gradually in all systems of the body. Morin hydrate (MH; 2',3,4',5,7-pentahydroxyflavone), a potent flavonoid abundantly present in plants of the Moraceae family, is thought to be a major bioactive compound that may be used to prevent a wide range of disease pathologies including hepatotoxicity. Therapeutic applications of morin (MOR) are however seriously constrained because of its insolubility, poor bioavailability, high metabolism and rapid elimination from the human body. Nanoformulation of MOR is a possible solution to these problems. In the present study we investigated the effectiveness of morin encapsulated chitosan nanoparticles (MCNPs) against arsenic induced liver damage in mice. MNCPs with an average diameter of 124.5 nm, a zeta potential of +16.2 mV and an encapsulation efficiency of 78% were prepared. Co-treatment of MOR and MCNPs by oral gavage on alternate days reduced the serum levels of AST, ALT, and ALP that were elevated in arsenic treated mice. The efficiency of MCNPs was found to be nearly 4 times higher than that of free MOR. Haematological and serum biochemical parameters including lipid profiles altered by arsenic were normalized following MCNP treatment. Arsenic deposition was lowered in the presence of MCNPs. Administration of MCNPs markedly inhibited ROS generation and elevated MDA levels in arsenic exposed mice. The level of hepatic antioxidant factors such as nuclear Nrf2 (Nrf2), superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), GSH peroxidase (GPx), glutathione-S-transferase (GST), heme oxygenase-1 (HO-1), and NADPH quinone oxidoreductase 1(NQO1) were markedly enhanced in the arsenic + MCNP group. Treatment by MCNPs prevented the arsenic induced damage of tissue histology. Also, MCNPs suppressed the arsenic induced pro- and anti-apoptotic parameters and attenuated the level of inflammatory mediators. Our data suggest that MCNPs are good hepatoprotective agents compared to free morin against arsenic induced toxicity and the protective effect results from its strong antioxidant, antiapoptotic and anti-inflammatory properties.
Collapse
Affiliation(s)
- Sanchaita Mondal
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology 4, Raja S.C. Mullick Road Kolkata-700032 West Bengal India
- Department of Chemistry, Jadavpur University 188, Raja S.C. Mullick Road Kolkata-700032 West Bengal India
| | - Sujata Das
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology 4, Raja S.C. Mullick Road Kolkata-700032 West Bengal India
| | - Pradip Kumar Mahapatra
- Department of Chemistry, Jadavpur University 188, Raja S.C. Mullick Road Kolkata-700032 West Bengal India
| | - Krishna Das Saha
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology 4, Raja S.C. Mullick Road Kolkata-700032 West Bengal India
| |
Collapse
|
280
|
Khan I, Baig MH, Mahfooz S, Imran MA, Khan MI, Dong JJ, Cho JY, Hatiboglu MA. Nanomedicine for Glioblastoma: Progress and Future Prospects. Semin Cancer Biol 2022; 86:172-186. [PMID: 35760272 DOI: 10.1016/j.semcancer.2022.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 06/09/2022] [Accepted: 06/21/2022] [Indexed: 11/29/2022]
Abstract
Glioblastoma is the most aggressive form of brain tumor, accounting for the highest mortality and morbidity rates. Current treatment for patients with glioblastoma includes maximal safe tumor resection followed by radiation therapy with concomitant temozolomide (TMZ) chemotherapy. The addition of TMZ to the conformal radiation therapy has improved the median survival time only from 12 months to 16 months in patients with glioblastoma. Despite these aggressive treatment strategies, patients' prognosis remains poor. This therapeutic failure is primarily attributed to the blood-brain barrier (BBB) that restricts the transport of TMZ from reaching the tumor site. In recent years, nanomedicine has gained considerable attention among researchers and shown promising developments in clinical applications, including the diagnosis, prognosis, and treatment of glioblastoma tumors. This review sheds light on the morphological and physiological complexity of the BBB. It also explains the development of nanomedicine strategies to enhance the permeability of drug molecules across the BBB.
Collapse
Affiliation(s)
- Imran Khan
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, Istanbul, Turkey
| | - Mohammad Hassan Baig
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Sadaf Mahfooz
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, Istanbul, Turkey
| | - Mohammad Azhar Imran
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Mohd Imran Khan
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Jae-June Dong
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Jae Yong Cho
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea.
| | - Mustafa Aziz Hatiboglu
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, Istanbul, Turkey; Department of Neurosurgery, Bezmialem Vakif University Medical School, Vatan Street, Fatih, Istanbul, Turkey.
| |
Collapse
|
281
|
Layer-By-Layer Self-Assembled Dip Coating for Antifouling Functionalized Finishing of Cotton Textile. Polymers (Basel) 2022; 14:polym14132540. [PMID: 35808585 PMCID: PMC9269539 DOI: 10.3390/polym14132540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 02/02/2023] Open
Abstract
The fouling of surfaces such as textiles is a major health challenge, and there is a continuous effort to develop materials and processes to overcome it. In consideration of this, this study regards the development of antifouling functional nanoencapsulated finishing for the cotton textile fabric by employing a layer-by-layer dip coating technique. Antifouling textile finishing was formulated by inducing the nanoencapsulation of the antifouling functional group inside the hydrophobic polymeric shell. Cotton fabric was taken as a substrate to incorporate antibacterial functionality by alternatively fabricating multilayers of antifouling polymeric formulation (APF) and polyelectrolyte solution. The surface morphology of nanoencapsulated finished textile fabric was characterized through scanning electron microscopy to confirm the uniform distribution of nanoparticles on the cotton textile fabric. Optical profilometry and atomic force microscopy studies indicated increased surface roughness in the coated textile substrate as compared to the uncoated textile. The surface thickness of the fabricated textile increased with the number of deposited bilayers on the textile substrate. Surface hydrophobicity increased with number of coating bilayers with θ values of x for single layer, up to y for 20 bilayers. The antibacterial activity of the uncoated and layer-by-layer coated finished textile was also evaluated. It was significant and exhibited a significant zone of inhibition against microbial strains Gram-positive S. aureus and Gram-negative E. coli. The bilayer coating exhibited water repellency, hydrophobicity, and antibacterial activity. Thus, the fabricated textile could be highly useful for many industrial and biomedical applications.
Collapse
|
282
|
Singh N, Handa M, Singh V, Kesharwani P, Shukla R. Lymphatic targeting for therapeutic application using nanoparticulate systems. J Drug Target 2022; 30:1017-1033. [PMID: 35722764 DOI: 10.1080/1061186x.2022.2092741] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The lymphatic system has grasped attention of researchers to a greater extent. The conventional methods of lymphatic delivery are now being modified to include nanotechnology to enhance the targeting of the drug at the specific pathological site. Scientists have worked successfully on different drug loaded nanocarriers that are modulated for the lymphatic system targeting for the treatment of various fatal diseases. Huge strides have been made in methods of delivery of these drugs either individually or in combination along with nanoparticles, therapeutic genes, and vaccines. However, the products introduced for commercial use are almost near nil. Altogether, there are challenges that need to be resolved and studies that are meant to be done for further improvements. The current review focuses on the understanding and pathophysiology of the lymphatic system and changes that occur during disease, drug characteristics, and physicochemical parameters that influence the lymphatic uptake of drugs and different nanocarriers. We further highlight different potential results obtained over the years with nanocarriers and other delivery methods to effectively target the lymphatic system for their therapeutic application. The challenges and drawbacks governing the lack of products available clinically have also been discussed.
Collapse
Affiliation(s)
- Nidhi Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P, India-226002
| | - Mayank Handa
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P, India-226002
| | - Vanshikha Singh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India-110062
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India-110062
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P, India-226002
| |
Collapse
|
283
|
Pirmardvand Chegini S, Varshosaz J, Dehghani A, Minaiyan M, Mirmohammad Sadeghi H. Ocular delivery of sunitinib-loaded nanoparticles doped in tragacanthic acid hydrogel in treatment of diabetic retinopathy in rats. Drug Dev Ind Pharm 2022; 48:29-39. [PMID: 35723593 DOI: 10.1080/03639045.2022.2092745] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Diabetic retinopathy (DR) is a common microvascular complication of diabetes mellitus. This study aimed to compare the effect of sunitinib-loaded poly (glycerol sebacate) (PGS)/gelatin nanoparticles doped in an injectable hydrogel with bevacizumab as a standard treatment of DR. METHODS The shear-sensitive hydrogel was prepared based on tragacanthic acid (TA) cross-linked with sodium acetate. DR was induced in rats by streptozotocin (STZ), and the animals were injected intravitreally a single dose of 20 µL sunitinib solution in three different concentrations (12.5, 25, and 50 µg/mL), sunitinib-loaded nanoparticles in hydrogel (413 μg/mL) and bevacizumab solution (6.25 mg/mL). The efficacy of the treatments was studied by histological and immunohisitological tests, angiogenesis, and optical coherence tomography (OCT). Vascular endothelial growth factor (VEGF) concentration was measured in the retina. RESULTS The results revealed that 20 µL of sunitinib with the concentration of 25 µg/mL was effective in DR without any disruption in the retina or any other side effects. This dose was considered the therapeutic dose for nanoparticles. Sunitinib loaded PGS/gelatine nanoparticles that were incorporated in the injectable hydrogel were as effective as bevacizumab in controlling DR. Although sunitinib solution reduced VEGF production and neovascularization in the retina compared to the negative control group, it was not as suitable as the nanoparticles. TA-based hydrogel showed no toxicity on the normal retina, and the angiography and histologic studies confirmed the VEGF results. Conclusions: Sunitinib nanoparticles doped in TA hydrogel may be an appropriate substitution of bevacizumab in the treatment of DR.
Collapse
Affiliation(s)
- Sana Pirmardvand Chegini
- Department of Pharmaceutics, Faculty of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jaleh Varshosaz
- Department of Pharmaceutics, Faculty of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Dehghani
- School of Medicine, Isfahan Eye Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Minaiyan
- Department of Pharmacology, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Mirmohammad Sadeghi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
284
|
Andrews J, Gkountouna O, Blaisten-Barojas E. Forecasting molecular dynamics energetics of polymers in solution from supervised machine learning. Chem Sci 2022; 13:7021-7033. [PMID: 35774160 PMCID: PMC9200117 DOI: 10.1039/d2sc01216b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/24/2022] [Indexed: 11/21/2022] Open
Abstract
Machine learning techniques including neural networks are popular tools for chemical, physical and materials applications searching for viable alternative methods in the analysis of structure and energetics of systems ranging from crystals to biomolecules. Efforts are less abundant for prediction of kinetics and dynamics. Here we explore the ability of three well established recurrent neural network architectures for reproducing and forecasting the energetics of a liquid solution of ethyl acetate containing a macromolecular polymer-lipid aggregate at ambient conditions. Data models from three recurrent neural networks, ERNN, LSTM and GRU, are trained and tested on half million points time series of the macromolecular aggregate potential energy and its interaction energy with the solvent obtained from molecular dynamics simulations. Our exhaustive analyses convey that the recurrent neural network architectures investigated generate data models that reproduce excellently the time series although their capability of yielding short or long term energetics forecasts with expected statistical distributions of the time points is limited. We propose an in silico protocol by extracting time patterns of the original series and utilizing these patterns to create an ensemble of artificial network models trained on an ensemble of time series seeded by the additional time patters. The energetics forecast improve, predicting a band of forecasted time series with a spread of values consistent with the molecular dynamics energy fluctuations span. Although the distribution of points from the band of energy forecasts is not optimal, the proposed in silico protocol provides useful estimates of the solvated macromolecular aggregate fate. Given the growing application of artificial networks in materials design, the data-based protocol presented here expands the realm of science areas where supervised machine learning serves as a decision making tool aiding the simulation practitioner to assess when long simulations are worth to be continued.
Collapse
Affiliation(s)
- James Andrews
- Center for Simulation and Modeling, George Mason University Fairfax Virginia 22030 USA
- Department of Computational and Data Sciences, George Mason University Fairfax Virginia 22030 USA
| | - Olga Gkountouna
- Department of Computational and Data Sciences, George Mason University Fairfax Virginia 22030 USA
| | - Estela Blaisten-Barojas
- Center for Simulation and Modeling, George Mason University Fairfax Virginia 22030 USA
- Department of Computational and Data Sciences, George Mason University Fairfax Virginia 22030 USA
| |
Collapse
|
285
|
Li W, Li S, Zhang J, Zhong H, Liang J, Huang S, Liao G, Zhang B, Liu C. Fabrication and evaluation of bone morphogenetic protein-2 microspheres coated black phosphorus nanosheets@polylactic-glycolic acid copolymers scaffold: A multifunctional antibacterial photothermal scaffold for bone regeneration. Int J Biol Macromol 2022; 210:350-364. [PMID: 35537585 DOI: 10.1016/j.ijbiomac.2022.05.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/25/2022] [Accepted: 05/04/2022] [Indexed: 12/25/2022]
Abstract
Clinical bone defects are often caused by high energy injury and are easily complicated by bacterial infection. An ideal bone repair material should promote bone regeneration and prevent bacterial infection. In this study, a multifunctional photothermal scaffold was developed: bone morphogenetic protein-2 (BMP-2)/polylactic-glycolic acid copolymers (PLGA) microspheres were prepared by a double emulsion method and then coated on the scaffolds prepared using a mixture of black phosphorus nanosheets (BPs) and PLGA, to form BMP-2@BPs scaffolds. The structural and photothermal properties of the composite scaffolds were characterized. The BMP-2@BPs scaffolds demonstrated good biocompatibility in both in vitro and in vivo experiments. The BMP-2@BPs scaffolds promoted osteogenic differentiation through a combination of BMP-2 release and upregulation of the expression of heat shock proteins by the radiation of near-infrared (NIR) light, which further upregulated the expression of osteogenesis-related genes. In addition, BPs demonstrated antibacterial effects under the mediation of NIR, which is beneficial for the prevention of clinical bacterial infections. In summary, the BMP-2@BPs scaffold was a multifunctional photothermal scaffold that could accelerate bone regeneration and act against bacteria. This study provides a new perspective for the treatment of bone defects and infectious bone defects.
Collapse
Affiliation(s)
- Wenhua Li
- Department of Trauma Orthopedics, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Avenue, Guangzhou 510280, Guangdong Province, China
| | - Siteng Li
- Department of Orthopaedic Trauma, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jinwei Zhang
- Department of Trauma Orthopedics, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Avenue, Guangzhou 510280, Guangdong Province, China
| | - Haoming Zhong
- Department of Trauma Orthopedics, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Avenue, Guangzhou 510280, Guangdong Province, China
| | - Jie Liang
- Department of Trauma Orthopedics, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Avenue, Guangzhou 510280, Guangdong Province, China
| | - Shijia Huang
- Department of Trauma Orthopedics, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Avenue, Guangzhou 510280, Guangdong Province, China
| | - Gaozu Liao
- School of Environment, South China Normal University, Higher Education Mega Center, Guangzhou 510006, China.
| | - Bao Zhang
- Three-level Biosafety Laboratory, School of Public Health, Southern Medical University, 1023 Sha Tai Nan Rd, Guangzhou 510080, China.
| | - Chenglong Liu
- Department of Trauma Orthopedics, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Avenue, Guangzhou 510280, Guangdong Province, China.
| |
Collapse
|
286
|
Nyambura CW, Sampath J, Nance E, Pfaendtner J. Exploring structure and dynamics of the polylactic‐co‐glycolic acid–polyethylene glycol copolymer and its homopolymer constituents in various solvents using all‐atom molecular dynamics. J Appl Polym Sci 2022. [DOI: 10.1002/app.52732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Chris W. Nyambura
- Department of Chemical Engineering University of Washington Seattle Washington USA
| | - Janani Sampath
- Department of Chemical Engineering University of Florida Gainesville Florida USA
| | - Elizabeth Nance
- Department of Chemical Engineering University of Washington Seattle Washington USA
| | - Jim Pfaendtner
- Department of Chemical Engineering University of Washington Seattle Washington USA
| |
Collapse
|
287
|
Gupta A, Sood A, Fuhrer E, Djanashvili K, Agrawal G. Polysaccharide-Based Theranostic Systems for Combined Imaging and Cancer Therapy: Recent Advances and Challenges. ACS Biomater Sci Eng 2022; 8:2281-2306. [PMID: 35513349 DOI: 10.1021/acsbiomaterials.1c01631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Designing novel systems for efficient cancer treatment and improving the quality of life for patients is a prime requirement in the healthcare sector. In this regard, theranostics have recently emerged as a unique platform, which combines the benefits of both diagnosis and therapeutics delivery. Theranostics have the desired contrast agent and the drugs combined in a single carrier, thus providing the opportunity for real-time imaging to monitor the therapy results. This helps in reducing the hazards related to treatment overdose or underdose and gives the possibility of personalized therapy. Polysaccharides, as natural biomolecules, have been widely explored to develop theranostics, as they act as a matrix for simultaneously loading both contrast agents and drugs for their utility in drug delivery and imaging. Additionally, their remarkable physicochemical attributes (biodegradability, satisfactory safety profile, abundance, and diversity in functionality and charge) can be tuned via postmodification, which offers numerous possibilities to develop theranostics with desired characteristics. Hence, we provide an overview of recent advances in polysaccharide matrix-based theranostics for drug delivery combined with magnetic resonance imaging, computed tomography, positron emission tomography, single photon emission computed tomography, and ultrasound imaging. Herein, we also summarize the toxicity assessment of polysaccharides, associated contrast agents, and nanotoxicity along with the challenges and future research directions.
Collapse
Affiliation(s)
- Aastha Gupta
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh-175075, India
| | - Ankur Sood
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh-175075, India
| | - Erwin Fuhrer
- School of Computing and Electrical Engineering, Indian Institute of Technology Mandi, Himachal Pradesh-175075, India
| | - Kristina Djanashvili
- Department of Biotechnology, Delft University of Technology, Van der Maasweg 9, 2629 HZ Delft, The Netherlands
| | - Garima Agrawal
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh-175075, India
| |
Collapse
|
288
|
Nanomaterials-Based Combinatorial Therapy as a Strategy to Combat Antibiotic Resistance. Antibiotics (Basel) 2022; 11:antibiotics11060794. [PMID: 35740200 PMCID: PMC9220075 DOI: 10.3390/antibiotics11060794] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 01/10/2023] Open
Abstract
Since the discovery of antibiotics, humanity has been able to cope with the battle against bacterial infections. However, the inappropriate use of antibiotics, the lack of innovation in therapeutic agents, and other factors have allowed the emergence of new bacterial strains resistant to multiple antibiotic treatments, causing a crisis in the health sector. Furthermore, the World Health Organization has listed a series of pathogens (ESKAPE group) that have acquired new and varied resistance to different antibiotics families. Therefore, the scientific community has prioritized designing and developing novel treatments to combat these ESKAPE pathogens and other emergent multidrug-resistant bacteria. One of the solutions is the use of combinatorial therapies. Combinatorial therapies seek to enhance the effects of individual treatments at lower doses, bringing the advantage of being, in most cases, much less harmful to patients. Among the new developments in combinatorial therapies, nanomaterials have gained significant interest. Some of the most promising nanotherapeutics include polymers, inorganic nanoparticles, and antimicrobial peptides due to their bactericidal and nanocarrier properties. Therefore, this review focuses on discussing the state-of-the-art of the most significant advances and concludes with a perspective on the future developments of nanotherapeutic combinatorial treatments that target bacterial infections.
Collapse
|
289
|
Nooreen R, Nene S, Jain H, Prasannanjaneyulu V, Chitlangya P, Otavi S, Khatri DK, Raghuvanshi RS, Singh SB, Srivastava S. Polymer nanotherapeutics: A versatile platform for effective rheumatoid arthritis therapy. J Control Release 2022; 348:397-419. [PMID: 35660632 DOI: 10.1016/j.jconrel.2022.05.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 11/27/2022]
Abstract
Rheumatoid arthritis is an aggressive and severely debilitating disorder that is characterized by joint pain and cartilage damage. It restricts mobility in patients, leaving them unable to carry out simple tasks. RA presents itself with severe lasting pain, swelling and stiffness in the joints and may cause permanent disability in patients. Treatment regimens currently employed for rheumatoid arthritis revolve around keeping clinical symptoms like joint pain, inflammation, swelling and stiffness at bay. The current therapeutic interventions in rheumatoid arthritis involve the use of non-steroidal anti-inflammatory drugs, glucocorticoids, disease-modifying anti-rheumatic drugs and newer biological drugs that are engineered for inhibiting the expression of pro-inflammatory mediators. These conventional drugs are plagued with severe adverse effects because of their higher systemic distribution, lack of specificity and higher doses. Oral, intra-articular, and intravenous routes are routinely used for drug delivery which is associated with decreased patient compliance, high cost, poor bioavailability and rapid systemic clearance. All these drawbacks have enticed researchers to create novel strategies for drug delivery, the main approach being nanocarrier-based systems. In this article, we aim to consolidate the remarkable contributions of polymeric carrier systems including microneedle technology and smart trigger-responsive polymeric carriers in the management of rheumatoid arthritis along with its detailed pathophysiology. This review also briefly describes the safety and regulatory aspects of polymer therapeutics.
Collapse
Affiliation(s)
- Rimsha Nooreen
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Shweta Nene
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Harsha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Velpula Prasannanjaneyulu
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Palak Chitlangya
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Shivam Otavi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Rajeev Singh Raghuvanshi
- Indian Pharmacopoeia Commission, Ministry of Health & Family Welfare, Government of India, India
| | - Shashi Bala Singh
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India.
| |
Collapse
|
290
|
Li X, Omonova Tuychi Qizi C, Mohamed Khamis A, Zhang C, Su Z. Nanotechnology for Enhanced Cytoplasmic and Organelle Delivery of Bioactive Molecules to Immune Cells. Pharm Res 2022; 39:1065-1083. [PMID: 35661086 DOI: 10.1007/s11095-022-03284-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/30/2022] [Indexed: 12/18/2022]
Abstract
Immune cells stand as a critical component of the immune system to maintain the internal environment homeostasis. The dysfunction of immune cells can result in various life-threatening diseases, including refractory infection, diabetes, cardiovascular disease, and cancer. Therefore, strategies to standardize or even enhance the function of immune cells are critical. Recently, nanotechnology has been highly researched and extensively applied for enhancing the cytoplasmic delivery of bioactive molecules to immune cells, providing efficient approaches to correct in vivo and in vitro dysfunction of immune cells. This review focuses on the technologies and challenges involved in improving endo-lysosomal escape, cytoplasmic release and organelle targeted delivery of different bioactive molecules in immune cells. Furthermore, it will elaborate on the broader vision of applying nanotechnology for treating immune cell-related diseases and constructing immune therapies and cytopharmaceuticals as potential treatments for diseases.
Collapse
Affiliation(s)
- Xiaoyu Li
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Charos Omonova Tuychi Qizi
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Amari Mohamed Khamis
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China.
| | - Zhigui Su
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
291
|
Fattahi R, Mohebichamkhorami F, Khani MM, Soleimani M, Hosseinzadeh S. Aspirin effect on bone remodeling and skeletal regeneration: Review article. Tissue Cell 2022; 76:101753. [PMID: 35180553 DOI: 10.1016/j.tice.2022.101753] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/21/2022] [Accepted: 02/06/2022] [Indexed: 12/21/2022]
Abstract
Bone tissues are one of the most complex tissues in the body that regenerate and repair themselves spontaneously under the right physiological conditions. Within the limitations of treating bone defects, mimicking tissue engineering through the recruitment of scaffolds, cell sources and growth factors, is strongly recommended. Aspirin is one of the non-steroidal anti-inflammatory drugs (NSAIDs) and has been used in clinical studies for many years due to its anti-coagulant effect. On the other hand, aspirin and other NSAIDs activate cytokines and some mediators in osteoclasts, osteoblasts and their progenitor cells in a defect area, thereby promoting bone regeneration. It also stimulates angiogenesis by increasing migration of endothelial cells and the newly developed vessels are of emergency in bone fracture repair. This review covers the role of aspirin in bone tissue engineering and also, highlights its chemical reactions, mechanisms, dosages, anti-microbial and angiogenesis activities.
Collapse
Affiliation(s)
- Roya Fattahi
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Mohebichamkhorami
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Khani
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Simzar Hosseinzadeh
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
292
|
Kulshrestha R, Singh A, Kumar P, Nair DS, Batra J, Mishra A, Dinda A. Nanoapproach targeting TGFβ1-Smad pathway and modulating lung microenvironment. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
293
|
Sabatelle RC, Liu R, Hung YP, Bressler E, Neal EJ, Martin A, Ekladious I, Grinstaff MW, Colson YL. Ultra-high drug loading improves nanoparticle efficacy against peritoneal mesothelioma. Biomaterials 2022; 285:121534. [PMID: 35487067 PMCID: PMC9881475 DOI: 10.1016/j.biomaterials.2022.121534] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 01/31/2023]
Abstract
Peritoneal mesothelioma is an aggressive disease with a median survival of under three years, due to a lack of effective treatment options. Mesothelioma is traditionally considered a "chemoresistant" tumor; however, low intratumoral drug levels coupled with the inability to administer high systemic doses suggests that therapeutic resistance may be due to poor drug delivery rather than inherent biology. While patient survival may improve with repetitive local intraperitoneal infusions of chemotherapy throughout the perioperative period, these regimens carry associated toxicities and significant peri-operative morbidity. To circumvent these issues, we describe ultra-high drug loaded nanoparticles (NPs) composed of a unique poly(1,2-glycerol carbonate)-graft-succinate-paclitaxel (PGC-PTX + PTX) conjugate. PGC-PTX + PTX NPs are cytotoxic, localize to tumor in vivo, and improve survival in a murine model of human peritoneal mesothelioma after a single intraperitoneal (IP) injection compared to multiple weekly doses of the clinically utilized formulation PTX-C/E. Given their unique pharmacokinetics, a second intraperitoneal dose of PGC-PTX + PTX NPs one month later more than doubles the overall survival compared to the clinical control (122 versus 58 days). These results validate the clinical potential of prolonged local paclitaxel to treat intracavitary malignancies such as mesothelioma using a tailored polymer-mediated nanoparticle formulation.
Collapse
Affiliation(s)
- Robert C. Sabatelle
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA
| | - Rong Liu
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Yin P. Hung
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Eric Bressler
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA
| | - Eliza J. Neal
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Andrew Martin
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA
| | - Iriny Ekladious
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA,Corresponding Authors: Mark W. Grinstaff, , Yolonda L. Colson,
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA,Corresponding Authors: Mark W. Grinstaff, , Yolonda L. Colson,
| |
Collapse
|
294
|
Allahyari M. PLGA Nanoparticles as an Efficient Platform in Protein Vaccines Against Toxoplasma gondii. Acta Parasitol 2022; 67:582-591. [PMID: 35013939 DOI: 10.1007/s11686-021-00499-w] [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: 04/02/2021] [Accepted: 11/22/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Toxoplasma gondii (T. gondii) as an obligatory intracellular is widespread all over the world and causes considerable concerns in immunocompromised patients by developing toxoplasmic encephalitis and in pregnancy because of serious consequences in the fetus. Although vaccination is the only approach to overcome toxoplasmosis, there is no commercially available human vaccine against T. gondii. PURPOSE The remarkable features of poly (lactic-co-glycolic acid) (PLGA) particles have brought up the application of PLGA as a promising vaccine delivery vehicle against T. gondii and other intracellular parasites. This review focuses on the application of the PLGA delivery system in the development of preventive vaccines against T. gondii. METHODS In this study, all required data were collected from articles indexed in English databases, including Scopus, PubMed, Web of Science, Science Direct, and Google Scholar. RESULT Immunity against T. gondii, characteristics of PLGA particles as a delivery vehicle, and all researches on particulate PLGA vaccines with different T. gondii antigens and DNA against were discussed and their efficacies in conferring protection against a lethal challenge based on increased survival or reduced brain cyst loads have been shown. CONCLUSION Although various levels of protection against lethal challenge have been achieved through PLGA-based vaccinations, there is still no complete protection against T. gondii infection. Surprisingly, the application of surface modifications of PLGA particles by mucoadhesive polymers, cationic agents, DCs (dendritic cells) targeting receptors, specialized membranous epithelial cells (M-cells), and co-delivery of the desired antigen along with toll-like receptor ligands would be a revolutionized vaccine strategy against T. gondii.
Collapse
Affiliation(s)
- Mojgan Allahyari
- Recombinant Protein Production Department, Production and Research Complex, Pasteur Institute of Iran, Karaj, Iran.
| |
Collapse
|
295
|
Jain M, Kumar S, Aswal VK, Al-Ghamdi AA, Kumar Kailasa S, Malek NI. Amino Acid Induced Self-Assembled Vesicles of Choline Oleate: pH responsive Nano-carriers for Targeted and Localized Delivery of Doxorubicin for Breast Cancer. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
296
|
Pandian SRK, Vijayakumar KK, Murugesan S, Kunjiappan S. Liposomes: An emerging carrier for targeting Alzheimer's and Parkinson's diseases. Heliyon 2022; 8:e09575. [PMID: 35706935 PMCID: PMC9189891 DOI: 10.1016/j.heliyon.2022.e09575] [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: 12/15/2021] [Revised: 03/19/2022] [Accepted: 05/23/2022] [Indexed: 11/19/2022] Open
Abstract
The function of the brain can be affected by various factors that include infection, tumor, and stroke. The major disorders reported with altered brain function are Alzheimer's disease (AD), Parkinson's disease (PD), dementia, brain cancer, seizures, mental disorders, and other movement disorders. The major barrier in treating CNS disease is the blood-brain barrier (BBB), which protects the brain from toxic molecules, and the cerebrospinal fluid (CSF) barrier, which separates blood from CSF. Brain endothelial cells and perivascular elements provide an integrated cellular barrier, the BBB, which hamper the invasion of molecules from the blood to the brain. Even though many drugs are available to treat neurological disorders, it fails to reach the desired site with the required concentration. In this purview, liposomes can carry required concentrations of molecules intracellular by diverse routes such as carrier-mediated transport and receptor-mediated transcytosis. Surface modification of liposomes enables them to deliver drugs to various brain cells, including neurons, astrocytes, oligodendrocytes, and microglia. The research studies supported the role of liposomes in delivering drugs across BBB and in reducing the pathogenesis of AD and PD. The liposomes were surface-functionalized with various molecules to reach the cells intricated with the AD or PD pathogenesis. The targeted and sustained delivery of drugs by liposomes is disturbed due to the antibody formation, renal clearance, accelerated blood clearance, and complement activation-related pseudoallergy (CARPA). Hence, this review will focus on the characteristics, surface functionalization, drug loading, and biodistribution of liposomes respective to AD and PD. In addition, the alternative strategies to overcome immunogenicity are discussed briefly.
Collapse
Affiliation(s)
- Sureshbabu Ram Kumar Pandian
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, 626126, Tamilnadu, India
| | - Kevin Kumar Vijayakumar
- Department of Molecular Microbiology, School of Biotechnology, Madurai Kamaraj University, Madurai, Tamilnadu, India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Vidya Vihar, Pilani, 333031, Rajasthan, India
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, 626126, Tamilnadu, India
| |
Collapse
|
297
|
Wang C, Yang X, Jiang Y, Qi L, Zhuge D, Xu T, Guo Y, Deng M, Zhang W, Tian D, Yin Q, Li L, Zhang Z, Wang Y, Yang GY, Chen Y, Tang Y. Targeted delivery of fat extract by platelet membrane-cloaked nanocarriers for the treatment of ischemic stroke. J Nanobiotechnology 2022; 20:249. [PMID: 35642036 PMCID: PMC9153102 DOI: 10.1186/s12951-022-01461-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/14/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Our previous studies suggest that human fat extract (FE) contains a variety of angiogenic factors and may provide an alternative treatment option for stroke. However, the therapeutic effect is largely limited due to its short half-life, and inaccurate targeting. RESULTS Herein, we leverage the targeting abilities of platelets (PLTs) to the lesion area of stroke and Arg-Gly-Asp (RGD) peptides to the angiogenic blood vessels to develop a biomimetic nanocarrier that capable of delivering FE precisely to treat stroke. The biomimetic nanocarriers are comprised of FE-encapsulated PLGA (poly(lactic-co-glycolic acid)) core enclosed by RGD peptides decorated plasma membrane of PLTs, namely RGD-PLT@PLGA-FE. We found that RGD-PLT@PLGA-FE not only targeted damaged and inflamed blood vessels but also achieved rapid accumulation in the lesion area of ischemic brain. In addition, RGD-PLT@PLGA-FE kept a sustained release behavior of FE at the lesion site, effectively increased its half-life and promoted angiogenesis and neurogenesis with delivering neurotrophic factors including BDNF, GDNF and bFGF to the brain, that ultimately resulted in blood flow increase and neurobehavioral recovery. CONCLUSIONS In conclusion, our study provides a new strategy to design a biomimetic system for FE delivery and it is a promising modality for stroke therapy.
Collapse
Affiliation(s)
- Cheng Wang
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Xuewei Yang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, 325027, China
| | - Yixu Jiang
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Lin Qi
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Deli Zhuge
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, 325027, China
| | - Tongtong Xu
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Yiyan Guo
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Mingwu Deng
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Dongyan Tian
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, 325027, China
| | - Qingqing Yin
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, 325027, China
| | - Li Li
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, 325027, China
| | - Zhijun Zhang
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Yongting Wang
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Guo-Yuan Yang
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Yijie Chen
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, 325027, China.
| | - Yaohui Tang
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China.
| |
Collapse
|
298
|
Kowalik A, Majerek M, Mrowiec K, Solich J, Faron-Górecka A, Woźnicka O, Dziedzicka-Wasylewska M, Łukasiewicz S. Dopamine D 2 and Serotonin 5-HT 1A Dimeric Receptor-Binding Monomeric Antibody scFv as a Potential Ligand for Carrying Drugs Targeting Selected Areas of the Brain. Biomolecules 2022; 12:749. [PMID: 35740874 PMCID: PMC9221303 DOI: 10.3390/biom12060749] [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: 05/13/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
Targeted therapy uses multiple ways of ensuring that the drug will be delivered to the desired site. One of these ways is an encapsulation of the drug and functionalization of the surface. Among the many molecules that can perform such a task, the present work focused on the antibodies of single-chain variable fragments (scFvs format). We studied scFv, which specifically recognizes the dopamine D2 and serotonin 5-HT1A receptor heteromers. The scFvD2-5-HT1A protein was analyzed biochemically and biologically, and the obtained results indicated that the antibody is properly folded and non-toxic and can be described as low-immunogenic. It is not only able to bind to the D2-5-HT1A receptor heteromer, but it also influences the cAMP signaling pathway and-when surfaced on nanogold particles-it can cross the blood-brain barrier in in vitro models. When administered to mice, it decreased locomotor activity, matching the effect induced by clozapine. Thus, we are strongly convinced that scFvD2-5-HT1A, which was a subject of the present investigation, is a promising targeting ligand with the potential for the functionalization of nanocarriers targeting selected areas of the brain.
Collapse
Affiliation(s)
- Agata Kowalik
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.K.); (M.M.); (K.M.); (M.D.-W.)
| | - Mateusz Majerek
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.K.); (M.M.); (K.M.); (M.D.-W.)
| | - Krzysztof Mrowiec
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.K.); (M.M.); (K.M.); (M.D.-W.)
| | - Joanna Solich
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Science, 31-343 Krakow, Poland; (J.S.); (A.F.-G.)
| | - Agata Faron-Górecka
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Science, 31-343 Krakow, Poland; (J.S.); (A.F.-G.)
| | - Olga Woźnicka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, 30-387 Krakow, Poland;
| | - Marta Dziedzicka-Wasylewska
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.K.); (M.M.); (K.M.); (M.D.-W.)
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Science, 31-343 Krakow, Poland; (J.S.); (A.F.-G.)
| | - Sylwia Łukasiewicz
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.K.); (M.M.); (K.M.); (M.D.-W.)
| |
Collapse
|
299
|
Alsmadi MM, AL-Daoud NM, Obaidat RM, Abu-Farsakh NA. Enhancing Atorvastatin In Vivo Oral Bioavailability in the Presence of Inflammatory Bowel Disease and Irritable Bowel Syndrome Using Supercritical Fluid Technology Guided by wbPBPK Modeling in Rat and Human. AAPS PharmSciTech 2022; 23:148. [PMID: 35585214 DOI: 10.1208/s12249-022-02302-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022] Open
Abstract
Inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS) are common disorders that can change the body's physiology and drugs pharmacokinetics. Solid dispersion (SD) preparation using supercritical fluid technology (SFT) has many advantages. Our study aimed to explore the effect of IBS and IBD on atorvastatin (ATV) pharmacokinetics, enhance ATV oral bioavailability (BCS II drug) using SFT, and analyze drug-disease-formulation interaction using a whole-body physiologically based pharmacokinetic (wbPBPK) model in rat and human. A novel ATV formulation was prepared using SFT and characterized in vitro and in vivo in healthy, IBS, and IBD rats. The resulting ATV plasma levels were analyzed using a combination of conventional and wbPBPK approaches. The novel formulation increased ATV solubility by 20-fold and resulted in a zero-order release of up to 95%. Both IBS and IBD increased ATV exposure after oral and intravenous administration by more than 30%. The novel SFT formulation increased ATV bioavailability by 28, 14, and 18% in control, IBD, and IBD rat groups and resulted in more consistent exposure as compared to raw ATV solution. Higher improvements in ATV bioavailability of more than 2-fold upon receiving the novel SFT formulation were predicted by the human wbPBPK model as compared to receiving the conventional tablets. Finally, the established wbPBPK model could describe ATV ADME in the presence of IBS and IBD after oral administration of raw ATV and using the novel SFT formula and can help scale the optimized ATV dosing regimens in the presence of IBS and IBD from rats to humans.
Collapse
|
300
|
Foglizzo V, Marchiò S. Nanoparticles as Physically- and Biochemically-Tuned Drug Formulations for Cancers Therapy. Cancers (Basel) 2022; 14:cancers14102473. [PMID: 35626078 PMCID: PMC9139219 DOI: 10.3390/cancers14102473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/26/2022] [Accepted: 05/13/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Conventional antitumor drugs have limitations, including poor water solubility and lack of targeting capability, with consequent non-specific distribution, systemic toxicity, and low therapeutic index. Nanotechnology promises to overcome these drawbacks by exploiting the physical properties of diverse nanocarriers that can be linked to moieties with binding selectivity for cancer cells. The use of nanoparticles as therapeutic formulations allows a targeted delivery and a slow, controlled release of the drug(s), making them tunable modules for applications in precision medicine. In addition, nanoparticles are also being developed as cancer vaccines, offering an opportunity to increase both cellular and humoral immunity, thus providing a new weapon to beat cancer. Abstract Malignant tumors originate from a combination of genetic alterations, which induce activation of oncogenes and inactivation of oncosuppressor genes, ultimately resulting in uncontrolled growth and neoplastic transformation. Chemotherapy prevents the abnormal proliferation of cancer cells, but it also affects the entire cellular network in the human body with heavy side effects. For this reason, the ultimate aim of cancer therapy remains to selectively kill cancer cells while sparing their normal counterparts. Nanoparticle formulations have the potential to achieve this aim by providing optimized drug delivery to a pathological site with minimal accumulation in healthy tissues. In this review, we will first describe the characteristics of recently developed nanoparticles and how their physical properties and targeting functionalization are exploited depending on their therapeutic payload, route of delivery, and tumor type. Second, we will analyze how nanoparticles can overcome multidrug resistance based on their ability to combine different therapies and targeting moieties within a single formulation. Finally, we will discuss how the implementation of these strategies has led to the generation of nanoparticle-based cancer vaccines as cutting-edge instruments for cancer immunotherapy.
Collapse
Affiliation(s)
- Valentina Foglizzo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Serena Marchiò
- Department of Oncology, University of Torino, 10060 Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
- Correspondence: ; Tel.: +39-01199333239
| |
Collapse
|