1
|
Zhang XJ, Jiang XY, Ma YL, Huang FY, Huang ZW. Encapsulating taurine into liposomes: A promising therapeutic for liver fibrosis. World J Gastroenterol 2024; 30:4509-4513. [DOI: 10.3748/wjg.v30.i41.4509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
We summarize the mechanism by which taurine (Tau) inhibits autophagy and induces iron apoptosis in hepatic stellate cells. Tau interacts with autophagy regulates multifunctional proteins, microtubule-associated protein 1 light chain 3 Beta, and autophagy-related gene 5 to inhibit autophagy, binds to ferritin heavy chain 1 and nuclear receptor coactivator 4 to trigger ferritin autophagy, and interacts with glutathione peroxidase 4 to promote iron apoptosis. There is a solid rationale for developing Tau-based therapies targeting autophagy and ferroptosis regulation. From a pharmaceutical point of view, there are certain requirements for Tau protein delivery systems, such as loading efficiency, stability, and targeting. Nanomaterials should also contain a hydrophilic motif similar to Tau to optimize loading efficiency. Since Tau is a hydrophilic molecule with high water solubility, liposomes, micelles, and amphiphilic polymer nanoparticles may represent a superior choice. The nanostructure of the liposome includes a water region and a lipid membrane to sequester hydrophilic and hydrophobic drugs, respectively, whereas Tau is expected to be loaded into the water region. In addition, a representative method of actively targeting hematopoietic stem cells is introduced. A Tau-based method for the treatment of liver fibrosis is proposed based on the formulation of common liposomes (lecithin plus cholesterol).
Collapse
Affiliation(s)
- Xue-Juan Zhang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong Province, China
| | - Xiao-Yi Jiang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong Province, China
| | - Yi-Lin Ma
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong Province, China
| | - Fei-Yi Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong Province, China
| | - Zheng-Wei Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong Province, China
| |
Collapse
|
2
|
Shafiei FS, Abroun S. Recent advancements in nanomedicine as a revolutionary approach to treating multiple myeloma. Life Sci 2024; 356:122989. [PMID: 39197575 DOI: 10.1016/j.lfs.2024.122989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024]
Abstract
Multiple myeloma, the second most common hematological malignancy, remains incurable with a 5-year survival rate of approximately 50 % and recurrence rates near 100 %, despite significant attempts to develop effective medicines. Therefore, there is a pressing demand in the medical field for innovative and more efficient treatments for MM. Currently, the standard approach for treating MM involves administering high-dose chemotherapy, which frequently correlates with improved results; however, one major limiting factor is the significant side effects of these medications. Furthermore, the strategies used to deliver medications to tumors limit their efficacy, whether by rapid clearance from circulation or an insufficient concentration in cancer cells. Cancer treatment has shifted from cytotoxic, nonspecific chemotherapy regimens to molecularly targeted, rationally developed drugs with improved efficacy and fewer side effects. Nanomedicines may provide an effective alternative way to avoid these limits by delivering drugs into the complicated bone marrow microenvironment and efficiently reaching myeloma cells. Putting drugs into nanoparticles can make their pharmacokinetic and pharmacodynamic profiles much better. This can increase the drug's effectiveness in tumors, extend its time in circulation in the blood, and lower its off-target toxicity. In this review, we introduce several criteria for the rational design of nanomedicine to achieve the best anti-tumoral therapeutic results. Next, we discuss recent advances in nanomedicine for MM therapy.
Collapse
Affiliation(s)
- Fatemeh Sadat Shafiei
- Department Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Biotechnology, Iranian Research Organization for Science & Technology (IROST), Tehran, Iran
| | - Saeid Abroun
- Department of Hematology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
3
|
Lalchandani DS, Chenkual L, Pate S, Kulhari U, Sahu BD, Chella N, Porwal PK. Folic acid-conjugated long circulating co-encapsulated atorvastatin and quercetin solid lipid nanoparticles: pharmacokinetics and biodistribution in rats. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:7188-7199. [PMID: 39314175 DOI: 10.1039/d4ay00821a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Background: Solid lipid nanoparticles (SLNs) have emerged as effective carriers for the simultaneous delivery of two drugs. Moreover, the surface modification of SLNs enhances their targetability and minimizes side effects, rendering them a promising and dynamic strategy for addressing various life-threatening diseases. The assessment of pharmacokinetic parameters is a critical aspect of this approach. In the present study, we report the development and validation of an LC-MS/MS-based bioanalytical method for the quantification of Atorvastatin (ATR) and Quercetin (QUER) encapsulated in folic acid-modified SLNs as a drug delivery system to estimate their pharmacokinetics and tissue distribution. Method: FA-SLNs were synthesized by amide linkage formation (carbodiimide reaction) and tested for their haemocompatibility. Further, an LC-MS/MS method was developed on a C18 (3 × 100 mm, 2.7 μm) column using 0.1% v/v formic acid in water and acetonitrile as the mobile phase with a 0.3 mL min-1 flow rate. For detection, analytes were ionized using an electron spray ionization (ESI) source in multiple reaction monitoring (MRM) mode. MRM for the ATR (559.0 → 440.2) m/z and IS (482.1 → 257.8) m/z in positive polarity, and QUER (301.9 → 151.0) m/z in negative polarity were optimized. Results: Pharmacokinetics studies demonstrated an increase in the half-lives of ATR and QUER of about 6.4-fold and 5.7-fold, respectively, from FA-SLN compared to pure drugs. Further, the active targeting facilitated by FA conjugation showed increased mean residence time (MRT) and decreased clearance time, resulting in long circulation time without the enhanced retention of drugs in the tissues of rats. These findings underscore the potential of FA-modified ATR and QUER-loaded SLNs as an advanced drug delivery strategy in improving the therapeutic outcomes.
Collapse
Affiliation(s)
- Dimple S Lalchandani
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Guwahati (NIPER-G), Changsari, Guwahati, Assam 781101, India.
| | - Laltanpuii Chenkual
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Guwahati (NIPER-G), Changsari, Guwahati, Assam 781101, India.
| | - Sonali Pate
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Guwahati (NIPER-G), Changsari, Guwahati, Assam 781101, India.
| | - Uttam Kulhari
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati (NIPER-G), Changsari, Guwahati, Assam 781101, India
| | - Bidya Dhar Sahu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati (NIPER-G), Changsari, Guwahati, Assam 781101, India
| | - Naveen Chella
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research-Guwahati (NIPER-G), Changsari, Guwahati, Assam 781101, India.
| | - Pawan Kumar Porwal
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Guwahati (NIPER-G), Changsari, Guwahati, Assam 781101, India.
| |
Collapse
|
4
|
Cirillo G, Cappello AR, Curcio M, Fiorillo M, Frattaruolo L, Avena P, Scorzafave L, Dolce V, Nicoletta FP, Iemma F. Novel CD44-Targeted Albumin Nanoparticles: An Innovative Approach to Improve Breast Cancer Treatment. Int J Mol Sci 2024; 25:10560. [PMID: 39408889 PMCID: PMC11477043 DOI: 10.3390/ijms251910560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
This study introduces novel CD44-targeted and redox-responsive nanoparticles (FNPs), proposed as doxorubicin (DOX) delivery devices for breast cancer. A cationized and redox-responsive Human Serum Albumin derivative was synthesized by conjugating Human Serum Albumin with cystamine moieties and then ionically complexing it with HA. The suitability of FNPs for cancer therapy was assessed through physicochemical measurements of size distribution (mean diameter of 240 nm), shape, and zeta potential (15.4 mV). Nanoparticles possessed high DOX loading efficiency (90%) and were able to trigger the drug release under redox conditions of the tumor environment (55% release after 2 h incubation). The use of the carrier increased the cytotoxic effect of DOX by targeting the CD44 protein. It was shown that, upon loading, the cytotoxic effect of DOX was enhanced in relation to CD44 protein expression in both 2D and 3D models. DOX@FNPs significantly decrease cellular metabolism by reducing both oxygen consumption and extracellular acidification rates. Moreover, they decrease the expression of proteins involved in the oxidative phosphorylation pathway, consequently reducing cellular viability and motility, as well as breast cancer stem cells and spheroid formation, compared to free DOX. This new formulation could become pioneering in reducing chemoresistance phenomena and increasing the specificity of DOX in breast cancer patients.
Collapse
Affiliation(s)
| | | | - Manuela Curcio
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende (CS), Italy; (G.C.); (A.R.C.); (L.F.); (P.A.); (L.S.); (F.P.N.); (F.I.)
| | - Marco Fiorillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende (CS), Italy; (G.C.); (A.R.C.); (L.F.); (P.A.); (L.S.); (F.P.N.); (F.I.)
| | | | | | | | | | | | | |
Collapse
|
5
|
Yeo S, Jung S, Kim H, Ahn JH, Hwang SJ. 4-Hexylresorcinol Loaded Solid Lipid Nanoparticles for Enhancing Anticancer Activity. Pharmaceuticals (Basel) 2024; 17:1296. [PMID: 39458937 PMCID: PMC11514591 DOI: 10.3390/ph17101296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Cancer is one of the most significant threats to human health. Following surgical excision, chemotherapy is an effective strategy against remaining cancer cells. 4-hexylresorcinol (4-HR) has anti-cancer properties and exhibits hydrophobicity-induced aggregation in the blood that has trouble with targeted tumor delivery and cellular uptake of the drug. The purpose of this study is to encapsulate 4-HR into solid lipid nanoparticles (SLNs) to enhance its anti-cancer effect by avoiding aggregation and facilitating cellular uptake. METHODS 4-HR SLNs were prepared via hot melt homogenization with sonication. SLN characteristics were assessed by analyzing particle size, zeta potential, and drug release. Cytotoxicity, as an indicator of the anti-cancer effect, was evaluated against HeLa (cervical cancer in humans), A549 (lung cancer in humans), and CT-26 (colon carcinoma in mice) cell lines. RESULTS Particle size ranged from 169.4 to 644.8 nm, and zeta potential ranged from -19.8 to -40.3 mV, which are conducive to cellular uptake. Entrapment efficiency (EE) of 4-HR was found to be 75.0-96.5%. The cytotoxicity of 4-HR-loaded SLNs demonstrated enhanced anti-cancer effects compared to pure 4-HR. The enhancement of anti-cancer effects depended on reduced particle size based on cellular uptake, the EE, and the cell type. CONCLUSIONS These findings imply that 4-HR-loaded SLN is a promising strategy for chemotherapy in cancer treatment.
Collapse
Affiliation(s)
- Sooho Yeo
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea;
| | - Sukkyun Jung
- Research Center of Barunbarum Co., Seoul 06776, Republic of Korea;
| | - Haneul Kim
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea;
| | - Jun-Hyun Ahn
- Department of Biopharmaceutical Engineering, Hannam University, 1646 Yuseongdae-ro, Yuseong-gu, Daejeon 34054, Republic of Korea;
| | - Sung-Joo Hwang
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea;
| |
Collapse
|
6
|
Xu K, Zhang Y, Cheng H, Chen W, Chen C, Zhang M, Song H, Wang F. Triple-negative breast cancer treatment with core-shell Magnetic@Platinium-Metal organic framework/epirubicin nano-platforms for chemo-photodynamic based combinational therapy: A review. Medicine (Baltimore) 2024; 103:e39845. [PMID: 39331917 PMCID: PMC11441927 DOI: 10.1097/md.0000000000039845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2024] Open
Abstract
The combination of chemotherapy and photodynamic therapy (PDT), enabled by core-shell nano-platforms, is a promising method to improve cancer therapy by overcoming hypoxia and boosting drug penetration in breast tumor. Core-shell magnetic (iron oxide: Fe3O4)@platinum-metal organic framework/epirubicin (abbreviated as M@Pt-MOF/EPI) nano-platform is considered an effective cancer therapeutic agent. Relatively small particle size, round shape, and specific response to pH, are the key features of these nanomaterials to be used as promising therapeutic agents. Chemotherapy and photodynamic therapy, when applied in addition to the anticancer effects of nanomaterials, further enhance the therapeutic efficacy. The extensive use, utilization, and efficacy of Core-Shell Magnetic@Platinium-Metal Organic Framework/epirubicin Nano-Platforms for chemo-photodynamic combination therapy in the treatment of several cancers, including triple-negative breast cancer, are examined in this in-depth investigation.
Collapse
Affiliation(s)
- Kangjie Xu
- Department of Central Laboratory, Binhai County People's Hospital, Yancheng, China
| | - Yanhua Zhang
- Department of Obstetrics and Gynecology, Binhai County People's Hospital, Yancheng, China
| | - Hui Cheng
- Department of General Surgery, Binhai County People's Hospital, Yancheng, China
| | - Weipeng Chen
- Department of General Surgery, Binhai County People's Hospital, Yancheng, China
| | - Cheng Chen
- Department of General Surgery, Binhai County People's Hospital, Yancheng, China
| | - Minglei Zhang
- Department of Oncology, Binhai County People's Hospital, Yancheng, China
| | - He Song
- Department of Rehabilitation Medicine, Kanda College of Nanjing Medical University, Lianyungang, China
| | - Feng Wang
- Department of General Surgery, Binhai County People's Hospital, Yancheng, China
| |
Collapse
|
7
|
Yeo S, Wu H, Yoon I, Kim HS, Song YK, Lee WK. Enhanced Photodynamic Therapy Efficacy through Solid Lipid Nanoparticle of Purpurin-18-N-Propylimide Methyl Ester for Cancer Treatment. Int J Mol Sci 2024; 25:10382. [PMID: 39408712 PMCID: PMC11477127 DOI: 10.3390/ijms251910382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Photodynamic therapy (PDT) is an innovative cancer treatment that utilizes light. When light irradiates, purpurin-18-N-propylimide methyl ester (P18 N PI ME) generates reactive oxygen species that destroy cancer cells. The hydrophobic nature of P18 N PI ME presents challenges regarding its aggregation in the body, which can affect its effectiveness. This study aimed to enhance the bioavailability and effectiveness of cancer treatment by synthesizing P18 N PI ME and formulating P18 N PI ME-loaded solid lipid nanoparticles (SLNs). The efficacy of PDT was estimated using the 1,3-diphenylisobenzofuran (DPBF) assay and photocytotoxicity tests on the HeLa (human cervical carcinoma) and A549 (human lung carcinoma) cell lines. The P18 N PI ME-loaded SLNs demonstrated particle sizes in the range of 158.59 nm to 248.43 nm and zeta potentials in the range of -15.97 mV to -28.73 mV. These SLNs exhibited sustained release of P18 N PI ME. DPBF analysis revealed enhanced PDT effects with SLNs containing P18 N PI ME compared with standalone P18 N PI MEs. Photocytotoxicity assays indicated toxicity under light irradiation but no toxicity in the dark. Furthermore, the smallest-sized formulation exhibited the most effective photodynamic activity. These findings indicate the potential of P18 N PI ME-loaded SLNs as promising strategies for PDT in cancer therapy.
Collapse
Affiliation(s)
- Sooho Yeo
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; (S.Y.); (H.-S.K.)
- Center for Nano Manufacturing and Department of Nanoscience and Engineering, Inje University, Gimhae 50834, Republic of Korea; (H.W.); (I.Y.)
| | - Huiqiang Wu
- Center for Nano Manufacturing and Department of Nanoscience and Engineering, Inje University, Gimhae 50834, Republic of Korea; (H.W.); (I.Y.)
| | - Il Yoon
- Center for Nano Manufacturing and Department of Nanoscience and Engineering, Inje University, Gimhae 50834, Republic of Korea; (H.W.); (I.Y.)
| | - Hye-Soo Kim
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; (S.Y.); (H.-S.K.)
| | - Young Kyu Song
- Research Center of Dr. i&B Co., Daejeon 34047, Republic of Korea
| | - Woo Kyoung Lee
- Center for Nano Manufacturing and Department of Nanoscience and Engineering, Inje University, Gimhae 50834, Republic of Korea; (H.W.); (I.Y.)
| |
Collapse
|
8
|
Adibifar A, Salimi M, Rostamkhani N, Karami Z, Agh-Atabay AH, Rostamizadeh K. Folic acid-conjugated bovine serum albumin-coated selenium-ZIF-8 core/shell nanoparticles for dual target-specific drug delivery in breast cancer. Drug Deliv Transl Res 2024:10.1007/s13346-024-01714-7. [PMID: 39317912 DOI: 10.1007/s13346-024-01714-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Methotrexate (MTX), a frequently used chemotherapeutic agent, has limited water solubility, leading to rapid clearance even in local injections. In the present study, we developed folic acid-conjugated BSA-stabilized selenium-ZIF-8 core/shell nanoparticles for targeted delivery of MTX to combat breast cancer. FT-IR, XRD, SEM, TEM, and elemental mapping analysis confirmed the successful formation of FA-BSA@MTX@Se@ZIF-8. The developed nano-DDS had a mean diameter, polydispersity index, and zeta potential of 254.8 nm, 0.17, and - 16.5 mV, respectively. The release behavior of MTX from the nanocarriers was pH-dependent, where the cumulative release percentage at pH 5.4 was higher than at pH 7.4. BSA significantly improved the blood compatibility of nanoparticles so that after modifying their surface with BSA, the percentage of hemolysis decreased from 12.67 to 5.12%. The loading of methotrexate in BSA@Se@ZIF-8 nanoparticles reduced its IC50 on 4T1 cells from 40.29 µg/mL to 16.54 µg/mL, and by conjugating folic acid on the surface, this value even decreased to 12.27 µg/mL. In vivo evaluation of the inhibitory effect in tumor-bearing mice showed that FA-BSA@MTX@Se@ZIF-8 caused a 2.8-fold reduction in tumor volume compared to the free MTX, which is due to the anticancer effect of selenium nanoparticles, the pH sensitivity of ZIF-8, and the presence of folic acid on the surface as a targeting agent. More importantly, histological studies and animal body weight monitoring confirmed that developed nano-DDS does not have significant organ toxicity. Taking together, the incorporation of chemotherapeutics in folic acid-conjugated BSA-stabilized selenium-ZIF-8 nanoparticles may hold a significant impact in the field of future tumor management.
Collapse
Affiliation(s)
- Arghavan Adibifar
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Biomaterial, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Maryam Salimi
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Biomaterial, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Neda Rostamkhani
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Biomaterial, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Zahra Karami
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran.
| | | | - Kobra Rostamizadeh
- Department of Pharmaceutical Biomaterial, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Psychiatry and Behavioral Sciences, Department of Pharmacology, School of Medicine, University of Washington, Seattle, WA, 98104, USA.
| |
Collapse
|
9
|
Hameed H, Faheem S, Younas K, Jamshaid M, Ereej N, Hameed A, Munir R, Khokhar R. A comprehensive review on lipid-based nanoparticles via nose to brain targeting as a novel approach. J Microencapsul 2024:1-34. [PMID: 39286884 DOI: 10.1080/02652048.2024.2404414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
The central nervous system (CNS) has been a chief concern for millions of people worldwide, and many therapeutic medications are unable to penetrate the blood-brain barrier. Advancements in nanotechnology have enabled safe, effective, and precise delivery of medications towards specific brain regions by utilising a nose-to-brain targeting route. This method reduces adverse effects, increases medication bioavailability, and facilitates mucociliary clearance while promoting accumulation of drug in the targeted brain region. Recent developments in lipid-based nanoparticles, for instance solid lipid nanoparticles (SLNs), liposomes, nanoemulsions, and nano-structured lipid carriers have been explored. SLNs are currently the most promising drug carrier system because of their capability of transporting drugs across the blood-brain barrier at the intended brain site. This approach offers higher efficacy, controlled drug delivery, target specificity, longer circulation time, and a reduction in toxicity through a biomimetic mechanism.
Collapse
Affiliation(s)
- Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Saleha Faheem
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Komel Younas
- Faculty of Pharmacy, University Paris Saclay, Orsay, France
| | - Muhammad Jamshaid
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Nelofer Ereej
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Anam Hameed
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Lahore, Pakistan
| | - Rabia Munir
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Rabia Khokhar
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| |
Collapse
|
10
|
Kazeminava F, Javanbakht S, Latifi Z, Rasoulzadehzali M, Abbaszadeh M, Alimohammadzadeh B, Mahdipour M, Fattahi A, Hamishehkar H, Adibag Z, Nouri M. Ultrasound-assisted encapsulating folic acid-based carbon quantum dots within breast cancer cell-derived exosomes as a co-receptors-mediated anticancer nanocarrier for enhanced breast cancer therapy. Sci Rep 2024; 14:16941. [PMID: 39043763 PMCID: PMC11266556 DOI: 10.1038/s41598-024-67934-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
The nonspecific nature of cancer drug delivery often results in substantial toxic side effects during treatments for breast cancer. To mitigate these negative outcomes, our approach involves loading methotrexate (MTX) within carbon quantum dots (CQDs) synthesized from folic acid, which are then enveloped in exosomal membranes obtained from breast cancer cells (Ex@MTX-CQDs). Analysis utilizing nanoparticle tracking techniques has demonstrated that these Ex@MTX-CQDs maintain the physical and biochemical properties of their exosomal precursors. The release profile of MTX indicated a restricted release percentage (less than 10%) under normal physiological conditions, which is contrasted by a more consistent release rate (approximately 65%) when emulating the conditions found within tumor tissues. The toxicological assessments have confirmed that the presence of exosomes combined with leftover folic acid significantly improves the delivery efficacy of MTX directly to the cancerous cells through the binding to folate and heparan sulfate proteoglycan receptors. This process results in increased disruption of the mitochondrial membrane potential and subsequently triggers apoptosis, ultimately leading to the destruction of cancerous cells. Our research could potentially contribute to the further innovation and application of nanocarriers derived from biological sources for the targeted treatment of breast cancer.
Collapse
Affiliation(s)
- Fahimeh Kazeminava
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Siamak Javanbakht
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Latifi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Mahmoud Abbaszadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Adibag
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Harisa GI, Bakheit AH, Alshehri S, Attia SM, Attia MSM. Chitosan-enclosed SLN improved SV-induced hepatocellular cell carcinoma death by modulation of IQGAP gene expression, JNK, and HDAC activities. Mol Biol Rep 2024; 51:824. [PMID: 39023688 DOI: 10.1007/s11033-024-09757-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a global life-threatening problem and therapeutic interventions are still encountered. IQGAP genes are involved in HCC oncogenesis. The modulatory effect of statins on the expression of IQGAP genes is still unclear. This study aims to study the effect of free SV and chitosan (CS) decorated simvastatin (SV) loaded solid lipid nanoparticles (C-SV-SLNs) on HCC mortality. METHODS AND RESULTS Plain, SV-SLN, and C-SV- SLN were prepared and characterized in terms of particle size (PS), zeta potential (ZP), and polydispersity index (PDI). The biosafety of different SLN was investigated using fresh erythrocytes, moreover, cytotoxicity was investigated using HepG2 cell lines. The effect of SLNs on IQGAPs gene expression as well as JNK, HDAC6, and HDAC8 activity was investigated using PCR and MOE-docking. The current results displayed that SV-SLNs have nanosized, negative ZP and are homogenous, CS decoration shifts the ZP of SLN into cationic ZP. Furthermore, all SLNs exhibited desirable biosafety in terms of no deleterious effect on erythrocyte integrity. SV solution and SV-SLN significantly increase the mortality of HepG2 compared to undertreated cells, however, the effect of SV-SLN is more pronounced compared to free SV. Remarkably, C-SV-SLN elicits high HepG2 cell mortality compared to free SV and SV-SLN. The treatment of HepG2 cells with SV solution, SV-SLN, or C-SV-SLN significantly upregulates the IQGAP2 gene with repression of IQGAP1 and IQGAP3 genes. MOE-docking studies revealed both SV and tenivastatin exhibit interactions with the active sites of JNK, HDAC6, and HDAC8. Moreover, tenivastatin exhibited greater interactions with magnesium and zinc compared to SV. CONCLUSIONS This research provides novel insights into the therapeutic potential of SV, SV-SLN and C-SV-SLNs in HCC treatment, modulating critical signaling cascades involving IQGAPs, JNK, and HDAC. The development of C-SV-SLNs presents a promising strategy for effective HCC therapy.
Collapse
Affiliation(s)
- Gamaleldin I Harisa
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia.
| | - Ahmed H Bakheit
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Samiyah Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed S M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
12
|
Sharifi M, Kamalabadi-Farahani M, Salehi M, Ebrahimi-Brough S, Alizadeh M. Recent perspectives on the synergy of mesenchymal stem cells with micro/nano strategies in peripheral nerve regeneration-a review. Front Bioeng Biotechnol 2024; 12:1401512. [PMID: 39050683 PMCID: PMC11266111 DOI: 10.3389/fbioe.2024.1401512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/19/2024] [Indexed: 07/27/2024] Open
Abstract
Despite the intrinsic repair of peripheral nerve injury (PNI), it is important to carefully monitor the process of peripheral nerve repair, as peripheral nerve regeneration is slow and incomplete in large traumatic lesions. Hence, mesenchymal stem cells (MSCs) with protective and regenerative functions are utilized in synergy with innovative micro/nano technologies to enhance the regeneration process of peripheral nerves. Nonetheless, as MSCs are assessed using standard regenerative criteria including sensory-motor indices, structural features, and morphology, it is challenging to differentiate between the protective and regenerative impacts of MSCs on neural tissue. This study aims to analyze the process of nerve regeneration, particularly the performance of MSCs with and without synergistic approaches. It also focuses on the paracrine secretions of MSCs and their conversion into neurons with functional properties that influence nerve regeneration after PNI. Furthermore, the study explores new ideas for nerve regeneration after PNI by considering the synergistic effect of MSCs and therapeutic compounds, neuronal cell derivatives, biological or polymeric conduits, organic/inorganic nanoparticles, and electrical stimulation. Finally, the study highlights the main obstacles to developing synergy in nerve regeneration after PNI and aims to open new windows based on recent advances in neural tissue regeneration.
Collapse
Affiliation(s)
- Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Kamalabadi-Farahani
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Majid Salehi
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Health Technology Incubator Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Somayeh Ebrahimi-Brough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Alizadeh
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
13
|
Kalra J, Baker J, Sun X, Kyle A, Minchinton A, Bally MB. Accumulation of liposomes in metastatic tumor sites is not necessary for anti-cancer drug efficacy. J Transl Med 2024; 22:621. [PMID: 38961395 PMCID: PMC11223361 DOI: 10.1186/s12967-024-05428-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND The tumor microenvironment is profoundly heterogeneous particularly when comparing sites of metastases. Establishing the extent of this heterogeneity may provide guidance on how best to design lipid-based drug delivery systems to treat metastatic disease. Building on our previous research, the current study employs a murine model of metastatic cancer to explore the distribution of ~ 100 nm liposomes. METHODS Female NCr nude mice were inoculated with a fluorescently labeled, Her2/neu-positive, trastuzumab-resistant breast cancer cell line, JIMT-1mkate, either in the mammary fat pad to create an orthotopic tumor (OT), or via intracardiac injection (IC) to establish tumors throughout the body. Animals were dosed with fluorescent and radio-labeled liposomes. In vivo and ex vivo fluorescent imaging was used to track liposome distribution over a period of 48 h. Liposome distribution in orthotopic tumors was compared to sites of tumor growth that arose following IC injection. RESULTS A significant amount of inter-vessel heterogeneity for DiR distribution was observed, with most tumor blood vessels showing little to no presence of the DiR-labelled liposomes. Further, there was limited extravascular distribution of DiR liposomes in the perivascular regions around DiR-positive vessels. While all OT tumors contained at least some DiR-positive vessels, many metastases had very little or none. Despite the apparent limited distribution of liposomes within metastases, two liposomal drug formulations, Irinophore C and Doxil, showed similar efficacy for both the OT and IC JIMT-1mkate models. CONCLUSION These findings suggest that liposomal formulations achieve therapeutic benefits through mechanisms that extend beyond the enhanced permeability and retention effect.
Collapse
Affiliation(s)
- Jessica Kalra
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada.
| | - Jennifer Baker
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - XuXin Sun
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Alastair Kyle
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Andrew Minchinton
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marcel B Bally
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- NanoMedicine Innovation Network, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
14
|
Raeispour S, Rahmandoust M, Kouchakzadeh H. A nanocarrier system based on CQDs for efficient mitoxantrone drug delivery. Heliyon 2024; 10:e31674. [PMID: 38841446 PMCID: PMC11152690 DOI: 10.1016/j.heliyon.2024.e31674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024] Open
Abstract
Cancer is the second most fatal disease among women. In recent years, utilizing strategies based on carbon quantum dots (CQDs) as targeted drug delivery systems has had a significant impact on advancing and improving cancer treatment. This study is focused on the development of a nanocarrier, based on CQDs, for improving the therapeutic efficiency of mitoxantrone (MTX). Hence, the N-doped CQDs were synthesized by a hydrothermal method. Following its purification, MTX was loaded to the CQD, resulting in an increase in the size from 36.78 ± 0.9 nm to 157.8 ± 12.18 nm, with an ideal drug entrapment efficiency of 97 %. Drug release investigation showed a pH-dependent improvement, from 8 % at pH 7.4 to 11 % at pH 5.2 after 48 h. Based on the Methylthiazolyldiphenyl-tetrazolium bromide (MTT) results after 5 h of treatment on MCF-7 breast cancer cells, the N-doped CQD showed no significant effect on the cancer cells, whereas a half maximal Inhibitory Concentration (IC50) was achieved with the N-doped CQD-MTX complex at a concentration between 0.5 to 0.8 μM. Therefore, the newly developed drug delivery complex was capable of providing a rather identical influence on MCF-7 cells, as the free MTX, however, improving the pharmacokinetic of the drug by its controlled and on-target drug release, due to an alteration in distribution and absorption parameters.
Collapse
Affiliation(s)
- Shahrzad Raeispour
- Protein Research Center, Shahid Beheshti University, Velenjak, 1983969411, Tehran, Iran
| | - Moones Rahmandoust
- Protein Research Center, Shahid Beheshti University, Velenjak, 1983969411, Tehran, Iran
| | - Hasan Kouchakzadeh
- Protein Research Center, Shahid Beheshti University, Velenjak, 1983969411, Tehran, Iran
| |
Collapse
|
15
|
Li J, He H, Liu S, Li X, Wu F. Revealing tumor cells and tissues with high selectivity through folic acid-targeted nanofluorescence probes responsive to acidic microenvironments. Front Oncol 2024; 14:1404148. [PMID: 38933449 PMCID: PMC11199542 DOI: 10.3389/fonc.2024.1404148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Tumor-specific fluorescent probes must fulfill the dual requirements of targeted accumulation within tumors and high-resolution imaging capabilities. To achieve both tumor-targeted accumulation and high-resolution imaging performance, we developed a composite comprising an acid-responsive bodipy conjugated to amphiphilic PEG-b-PLA polymer, along with folic acid (FA)-modified PEG-b-PLA as a targeting moiety for active tumor-specific accumulation. Finally, a novel assembly of hybrid fluorescent nanoparticles was successfully synthesized by integrating these two components, demonstrating exceptional responsiveness to acidic conditions for fluorescence excitation and remarkable tumor-targeted accumulation capabilities. We conducted comprehensive in vitro and in vivo investigations employing techniques such as analysis of physicochemical properties, fluorescence-based probes detection at varying pH levels, assessment of in vitro cytotoxicity, evaluation of cellular uptake capacity, analysis of lysosomal co-localization imaging, examination of tumor fluorescence images in vivo, and investigation of biological distribution patterns. The results demonstrated that the acid-responsive nanofluorescence probe we designed and synthesized possesses desirable physical and chemical characteristics, including a small particle size and low cytotoxicity. Moreover, it exhibits rapid real-time response to acidic environments and displays enhanced fluorescence intensity, enabling the real-time tracking of probe entry into tumor cells as well as intracellular lysozyme accumulation. We achieved highly specific in vivo tumor visualization by combining nanoprobes targeting folate receptor. Through imaging cervical tumor mice, we demonstrated the precise imaging performance and high targeted accumulation of FA-targeted nanofluorescence probes in tumor tissue. Furthermore, we confirmed the in vivo safety of the FA-targeted nanofluorescence probe through biological distribution analysis. These findings highlight the potential widespread application of FA-targeted acid-responsive nanofluorescence probes for selective imaging of tumor cells and tissues.
Collapse
Affiliation(s)
- Jing Li
- Neurobiology Laboratory, Wannan Medical College, Wuhu, China
| | - Hongyi He
- College of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Shuyan Liu
- Department of Obstetrics and Gynecology, Second Hospital of Jilin University, Changchun, China
| | - Xining Li
- School of Medicine, Huzhou University, Huzhou, China
| | - Fengfeng Wu
- Department of Orthopedics and Rehabilitation, Huzhou Hospital of Zhejiang University School of Medicine, Huzhou, China
- Department of Orthopedics and Rehabilitation, Huzhou Central Hospital, Huzhou, China
| |
Collapse
|
16
|
Naser IH, Zaid M, Ali E, Jabar HI, Mustafa AN, Alubiady MHS, Ramadan MF, Muzammil K, Khalaf RM, Jalal SS, Alawadi AH, Alsalamy A. Unveiling innovative therapeutic strategies and future trajectories on stimuli-responsive drug delivery systems for targeted treatment of breast carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3747-3770. [PMID: 38095649 DOI: 10.1007/s00210-023-02885-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/02/2023] [Indexed: 05/23/2024]
Abstract
This comprehensive review delineates the latest advancements in stimuli-responsive drug delivery systems engineered for the targeted treatment of breast carcinoma. The manuscript commences by introducing mammary carcinoma and the current therapeutic methodologies, underscoring the urgency for innovative therapeutic strategies. Subsequently, it elucidates the logic behind the employment of stimuli-responsive drug delivery systems, which promise targeted drug administration and the minimization of adverse reactions. The review proffers an in-depth analysis of diverse types of stimuli-responsive systems, including thermoresponsive, pH-responsive, and enzyme-responsive nanocarriers. The paramount importance of material choice, biocompatibility, and drug loading strategies in the design of these systems is accentuated. The review explores characterization methodologies for stimuli-responsive nanocarriers and probes preclinical evaluations of their efficacy, toxicity, pharmacokinetics, and biodistribution in mammary carcinoma models. Clinical applications of stimuli-responsive systems, ongoing clinical trials, the potential of combination therapies, and the utility of multifunctional nanocarriers for the co-delivery of assorted drugs and therapies are also discussed. The manuscript addresses the persistent challenge of drug resistance in mammary carcinoma and the potential of stimuli-responsive systems in surmounting it. Regulatory and safety considerations, including FDA guidelines and biocompatibility assessments, are outlined. The review concludes by spotlighting future trajectories and emergent technologies in stimuli-responsive drug delivery, focusing on pioneering approaches, advancements in nanotechnology, and personalized medicine considerations. This review aims to serve as a valuable compendium for researchers and clinicians interested in the development of efficacious and safe stimuli-responsive drug delivery systems for the treatment of breast carcinoma.
Collapse
Affiliation(s)
- Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Muhaned Zaid
- Department of Pharmacy, Al-Manara College for Medical Sciences, Maysan, Amarah, Iraq
| | - Eyhab Ali
- Al-Zahraa University for Women, Karbala, Iraq
| | - Hayder Imad Jabar
- Department of Pharmaceutics, College of Pharmacy, University of Al-Ameed, Karbala, Iraq
| | | | | | | | - Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University, Abha, Saudi Arabia
| | | | - Sarah Salah Jalal
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Ahmed Hussien Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq
- College of Technical Engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, the Islamic University of Babylon, Babylon, Iraq
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq.
| |
Collapse
|
17
|
Kalinova R, Videv P, Petrova S, Doumanov J, Dimitrov I. Poly(2-(dimethylamino)ethyl methacrylate)-Grafted Amphiphilic Block Copolymer Micelles Co-Loaded with Quercetin and DNA. Molecules 2024; 29:2540. [PMID: 38893415 PMCID: PMC11173910 DOI: 10.3390/molecules29112540] [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: 04/29/2024] [Revised: 05/17/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
The synergistic effect of drug and gene delivery is expected to significantly improve cancer therapy. However, it is still challenging to design suitable nanocarriers that are able to load simultaneously anticancer drugs and nucleic acids due to their different physico-chemical properties. In the present work, an amphiphilic block copolymer comprising a biocompatible poly(ethylene glycol) (PEG) block and a multi-alkyne-functional biodegradable polycarbonate (PC) block was modified with a number of poly(2-(dimethylamino)ethyl methacrylate) (PDMAEMA) side chains applying the highly efficient azide-alkyne "click" chemistry reaction. The resulting cationic amphiphilic copolymer with block and graft architecture (MPEG-b-(PC-g-PDMAEMA)) self-associated in aqueous media into nanosized micelles which were loaded with the antioxidant, anti-inflammatory, and anticancer drug quercetin. The drug-loaded nanoparticles were further used to form micelleplexes in aqueous media through electrostatic interactions with DNA. The obtained nanoaggregates-empty and drug-loaded micelles as well as the micelleplexes intended for simultaneous DNA and drug codelivery-were physico-chemically characterized. Additionally, initial in vitro evaluations were performed, indicating the potential application of the novel polymer nanocarriers as drug delivery systems.
Collapse
Affiliation(s)
- Radostina Kalinova
- Institute of Polymers, Bulgarian Academy of Sciences, Akad. G. Bonchev St., Bl. 103-A, 1113 Sofia, Bulgaria
| | - Pavel Videv
- Department of Biochemistry, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria; (P.V.); (S.P.); (J.D.)
| | - Svetla Petrova
- Department of Biochemistry, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria; (P.V.); (S.P.); (J.D.)
| | - Jordan Doumanov
- Department of Biochemistry, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria; (P.V.); (S.P.); (J.D.)
| | - Ivaylo Dimitrov
- Institute of Polymers, Bulgarian Academy of Sciences, Akad. G. Bonchev St., Bl. 103-A, 1113 Sofia, Bulgaria
| |
Collapse
|
18
|
Reznik I, Kolesova E, Pestereva A, Baranov K, Osin Y, Bogdanov K, Swart J, Moshkalev S, Orlova A. Synthesis of Submicron CaCO 3 Particles in 3D-Printed Microfluidic Chips Supporting Advection and Diffusion Mixing. MICROMACHINES 2024; 15:652. [PMID: 38793225 PMCID: PMC11123073 DOI: 10.3390/mi15050652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/02/2024] [Accepted: 05/05/2024] [Indexed: 05/26/2024]
Abstract
Microfluidic technology provides a solution to the challenge of continuous CaCO3 particle synthesis. In this study, we utilized a 3D-printed microfluidic chip to synthesize CaCO3 micro- and nanoparticles in vaterite form. Our primary focus was on investigating a continuous one-phase synthesis method tailored for the crystallization of these particles. By employing a combination of confocal and scanning electron microscopy, along with Raman spectroscopy, we were able to thoroughly evaluate the synthesis efficiency. This evaluation included aspects such as particle size distribution, morphology, and polymorph composition. The results unveiled the existence of two distinct synthesis regimes within the 3D-printed microfluidic chips, which featured a channel cross-section of 2 mm2. In the first regime, which was characterized by chaotic advection, particles with an average diameter of around 2 μm were produced, thereby displaying a broad size distribution. Conversely, the second regime, marked by diffusion mixing, led to the synthesis of submicron particles (approximately 800-900 nm in diameter) and even nanosized particles (70-80 nm). This research significantly contributes valuable insights to both the understanding and optimization of microfluidic synthesis processes, particularly in achieving the controlled production of submicron and nanoscale particles.
Collapse
Affiliation(s)
- Ivan Reznik
- International Research and Education Center for Physics of Nanostructures, ITMO University, Saint Petersburg 197101, Russia; (E.K.); (K.B.)
- Faculty of Electrical Engineering and Computing, University of Campinas, Campinas 13083-970, Brazil;
| | - Ekaterina Kolesova
- International Research and Education Center for Physics of Nanostructures, ITMO University, Saint Petersburg 197101, Russia; (E.K.); (K.B.)
- Research Center for Translation Medicine, Sirius University, Sochi 354349, Russia
| | - Anna Pestereva
- International Laboratory Hybrid Nanostructures for Biomedicine, ITMO University, Saint Petersburg 199034, Russia; (A.P.); (K.B.); (A.O.)
| | - Konstantin Baranov
- International Laboratory Hybrid Nanostructures for Biomedicine, ITMO University, Saint Petersburg 199034, Russia; (A.P.); (K.B.); (A.O.)
| | - Yury Osin
- Laboratory for Scientific Restoration of Precious Metals, The State Hermitage Museum, Saint Petersburg 191186, Russia;
| | - Kirill Bogdanov
- International Research and Education Center for Physics of Nanostructures, ITMO University, Saint Petersburg 197101, Russia; (E.K.); (K.B.)
| | - Jacobus Swart
- Faculty of Electrical Engineering and Computing, University of Campinas, Campinas 13083-970, Brazil;
| | - Stanislav Moshkalev
- Center for Semiconductor Components and Nanotechnology, University of Campinas, Campinas 13083-870, Brazil;
| | - Anna Orlova
- International Laboratory Hybrid Nanostructures for Biomedicine, ITMO University, Saint Petersburg 199034, Russia; (A.P.); (K.B.); (A.O.)
| |
Collapse
|
19
|
Urbano-Gámez JD, Guzzi C, Bernal M, Solivera J, Martínez-Zubiaurre I, Caro C, García-Martín ML. Tumor versus Tumor Cell Targeting in Metal-Based Nanoparticles for Cancer Theranostics. Int J Mol Sci 2024; 25:5213. [PMID: 38791253 PMCID: PMC11121233 DOI: 10.3390/ijms25105213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The application of metal-based nanoparticles (mNPs) in cancer therapy and diagnostics (theranostics) has been a hot research topic since the early days of nanotechnology, becoming even more relevant in recent years. However, the clinical translation of this technology has been notably poor, with one of the main reasons being a lack of understanding of the disease and conceptual errors in the design of mNPs. Strikingly, throughout the reported studies to date on in vivo experiments, the concepts of "tumor targeting" and "tumor cell targeting" are often intertwined, particularly in the context of active targeting. These misconceptions may lead to design flaws, resulting in failed theranostic strategies. In the context of mNPs, tumor targeting can be described as the process by which mNPs reach the tumor mass (as a tissue), while tumor cell targeting refers to the specific interaction of mNPs with tumor cells once they have reached the tumor tissue. In this review, we conduct a critical analysis of key challenges that must be addressed for the successful targeting of either tumor tissue or cancer cells within the tumor tissue. Additionally, we explore essential features necessary for the smart design of theranostic mNPs, where 'smart design' refers to the process involving advanced consideration of the physicochemical features of the mNPs, targeting motifs, and physiological barriers that must be overcome for successful tumor targeting and/or tumor cell targeting.
Collapse
Affiliation(s)
- Jesús David Urbano-Gámez
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - Cinzia Guzzi
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - Manuel Bernal
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, 29071 Malaga, Spain
| | - Juan Solivera
- Department of Neurosurgery, Reina Sofia University Hospital, 14004 Cordoba, Spain;
| | - Iñigo Martínez-Zubiaurre
- Department of Clinical Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, P.O. Box 6050, Langnes, 9037 Tromsö, Norway;
| | - Carlos Caro
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - María Luisa García-Martín
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
- Biomedical Research Networking Center in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
20
|
Beach M, Nayanathara U, Gao Y, Zhang C, Xiong Y, Wang Y, Such GK. Polymeric Nanoparticles for Drug Delivery. Chem Rev 2024; 124:5505-5616. [PMID: 38626459 PMCID: PMC11086401 DOI: 10.1021/acs.chemrev.3c00705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
The recent emergence of nanomedicine has revolutionized the therapeutic landscape and necessitated the creation of more sophisticated drug delivery systems. Polymeric nanoparticles sit at the forefront of numerous promising drug delivery designs, due to their unmatched control over physiochemical properties such as size, shape, architecture, charge, and surface functionality. Furthermore, polymeric nanoparticles have the ability to navigate various biological barriers to precisely target specific sites within the body, encapsulate a diverse range of therapeutic cargo and efficiently release this cargo in response to internal and external stimuli. However, despite these remarkable advantages, the presence of polymeric nanoparticles in wider clinical application is minimal. This review will provide a comprehensive understanding of polymeric nanoparticles as drug delivery vehicles. The biological barriers affecting drug delivery will be outlined first, followed by a comprehensive description of the various nanoparticle designs and preparation methods, beginning with the polymers on which they are based. The review will meticulously explore the current performance of polymeric nanoparticles against a myriad of diseases including cancer, viral and bacterial infections, before finally evaluating the advantages and crucial challenges that will determine their wider clinical potential in the decades to come.
Collapse
Affiliation(s)
- Maximilian
A. Beach
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Umeka Nayanathara
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yanting Gao
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Changhe Zhang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yijun Xiong
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yufu Wang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Georgina K. Such
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
21
|
Cornelius P, Mayes BA, Petersen JS, Turnquist DJ, Dufour PJ, Dannenberg AJ, Shanahan JM, Carver BJ. Pharmacological Characterization of SDX-7320/Evexomostat: A Novel Methionine Aminopeptidase Type 2 Inhibitor with Anti-tumor and Anti-metastatic Activity. Mol Cancer Ther 2024; 23:595-605. [PMID: 38530115 PMCID: PMC11063762 DOI: 10.1158/1535-7163.mct-23-0574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/07/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024]
Abstract
Methionine aminopeptidase type 2 (METAP2) is a ubiquitous, evolutionarily conserved metalloprotease fundamental to protein biosynthesis which catalyzes removal of the N-terminal methionine residue from nascent polypeptides. METAP2 is an attractive target for cancer therapeutics based upon its over-expression in multiple human cancers, the importance of METAP2-specific substrates whose biological activity may be altered following METAP2 inhibition, and additionally, that METAP2 was identified as the target for the anti-angiogenic natural product, fumagillin. Irreversible inhibition of METAP2 using fumagillin analogues has established the anti-angiogenic and anti-tumor characteristics of these derivatives; however, their full clinical potential has not been realized due to a combination of poor drug-like properties and dose-limiting central nervous system (CNS) toxicity. This report describes the physicochemical and pharmacological characterization of SDX-7320 (evexomostat), a polymer-drug conjugate of the novel METAP2 inhibitor (METAP2i) SDX-7539. In vitro binding, enzyme, and cell-based assays demonstrated that SDX-7539 is a potent and selective METAP2 inhibitor. In utilizing a high molecular weight, water-soluble polymer to conjugate the novel fumagillol-derived, cathepsin-released, METAP2i SDX-7539, limitations observed with prior generation, small molecule fumagillol derivatives were ameliorated including reduced CNS exposure of the METAP2i, and prolonged half-life enabling convenient administration. Multiple xenograft and syngeneic cancer models were utilized to demonstrate the anti-tumor and anti-metastatic profile of SDX-7320. Unlike polymer-drug conjugates in general, reductions in small molecule-equivalent efficacious doses following polymer conjugation were observed. SDX-7320 has completed a phase I clinical safety study in patients with late-stage cancer and is currently being evaluated in multiple phase Ib/II clinical studies in patients with advanced solid tumors.
Collapse
|
22
|
Deleuziere M, Benoist É, Quelven I, Gras E, Amiens C. [ 18F]-Radiolabelled Nanoplatforms: A Critical Review of Their Intrinsic Characteristics, Radiolabelling Methods, and Purification Techniques. Molecules 2024; 29:1537. [PMID: 38611815 PMCID: PMC11013168 DOI: 10.3390/molecules29071537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
A wide range of nano-objects is found in many applications of our everyday life. Recognition of their peculiar properties and ease of functionalization has prompted their engineering into multifunctional platforms that are supposed to afford efficient tools for the development of biomedical applications. However, bridging the gap between bench to bedside cannot be expected without a good knowledge of their behaviour in vivo, which can be obtained through non-invasive imaging techniques, such as positron emission tomography (PET). Their radiolabelling with [18F]-fluorine, a technique already well established and widely used routinely for PET imaging, with [18F]-FDG for example, and in preclinical investigation using [18F]-radiolabelled biological macromolecules, has, therefore, been developed. In this context, this review highlights the various nano-objects studied so far, the reasons behind their radiolabelling, and main in vitro and/or in vivo results obtained thereof. Then, the methods developed to introduce the radioelement are presented. Detailed indications on the chemical steps involved are provided, and the stability of the radiolabelling is discussed. Emphasis is then made on the techniques used to purify and analyse the radiolabelled nano-objects, a point that is rarely discussed despite its technical relevance and importance for accurate imaging. The pros and cons of the different methods developed are finally discussed from which future work can develop.
Collapse
Affiliation(s)
- Maëlle Deleuziere
- SPCMIB, CNRS UMR 5068, Université de Toulouse III Paul Sabatier, 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (M.D.); (É.B.)
- Toulouse NeuroImaging Center (ToNIC), INSERM/UPS UMR 1214, University Hospital of Toulouse-Purpan, CEDEX 3, 31024 Toulouse, France;
- LCC-CNRS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | - Éric Benoist
- SPCMIB, CNRS UMR 5068, Université de Toulouse III Paul Sabatier, 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (M.D.); (É.B.)
| | - Isabelle Quelven
- Toulouse NeuroImaging Center (ToNIC), INSERM/UPS UMR 1214, University Hospital of Toulouse-Purpan, CEDEX 3, 31024 Toulouse, France;
| | - Emmanuel Gras
- Laboratoire Hétérochimie Fondamentale et Appliquée, UMR 5069, CNRS—Université de Toulouse, 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France;
| | - Catherine Amiens
- LCC-CNRS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| |
Collapse
|
23
|
Negut I, Bita B. Polymersomes as Innovative, Stimuli-Responsive Platforms for Cancer Therapy. Pharmaceutics 2024; 16:463. [PMID: 38675124 PMCID: PMC11053450 DOI: 10.3390/pharmaceutics16040463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
This review addresses the urgent need for more targeted and less toxic cancer treatments by exploring the potential of multi-responsive polymersomes. These advanced nanocarriers are engineered to deliver drugs precisely to tumor sites by responding to specific stimuli such as pH, temperature, light, hypoxia, and redox conditions, thereby minimizing the side effects associated with traditional chemotherapy. We discuss the design, synthesis, and recent applications of polymersomes, emphasizing their ability to improve therapeutic outcomes through controlled drug release and targeted delivery. Moreover, we highlight the critical areas for future research, including the optimization of polymersome-biological interactions and biocompatibility, to facilitate their clinical adoption. Multi-responsive polymersomes emerge as a promising development in nanomedicine, offering a pathway to safer and more effective cancer treatments.
Collapse
Affiliation(s)
- Irina Negut
- Faculty of Physics, University of Bucharest, 077125 Magurele, Romania;
| | - Bogdan Bita
- Faculty of Physics, University of Bucharest, 077125 Magurele, Romania;
- National Institute for Lasers, Plasma and Radiation Physics, 077125 Magurele, Romania
| |
Collapse
|
24
|
Cooley MB, Wegierak D, Exner AA. Using imaging modalities to predict nanoparticle distribution and treatment efficacy in solid tumors: The growing role of ultrasound. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1957. [PMID: 38558290 PMCID: PMC11006412 DOI: 10.1002/wnan.1957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 12/22/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
Nanomedicine in oncology has not had the success in clinical impact that was anticipated in the early stages of the field's development. Ideally, nanomedicines selectively accumulate in tumor tissue and reduce systemic side effects compared to traditional chemotherapeutics. However, this has been more successful in preclinical animal models than in humans. The causes of this failure to translate may be related to the intra- and inter-patient heterogeneity of the tumor microenvironment. Predicting whether a patient will respond positively to treatment prior to its initiation, through evaluation of characteristics like nanoparticle extravasation and retention potential in the tumor, may be a way to improve nanomedicine success rate. While there are many potential strategies to accomplish this, prediction and patient stratification via noninvasive medical imaging may be the most efficient and specific strategy. There have been some preclinical and clinical advances in this area using MRI, CT, PET, and other modalities. An alternative approach that has not been studied as extensively is biomedical ultrasound, including techniques such as multiparametric contrast-enhanced ultrasound (mpCEUS), doppler, elastography, and super-resolution processing. Ultrasound is safe, inexpensive, noninvasive, and capable of imaging the entire tumor with high temporal and spatial resolution. In this work, we summarize the in vivo imaging tools that have been used to predict nanoparticle distribution and treatment efficacy in oncology. We emphasize ultrasound imaging and the recent developments in the field concerning CEUS. The successful implementation of an imaging strategy for prediction of nanoparticle accumulation in tumors could lead to increased clinical translation of nanomedicines, and subsequently, improved patient outcomes. This article is categorized under: Diagnostic Tools In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery Emerging Technologies.
Collapse
Affiliation(s)
- Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana Wegierak
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Radiology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| |
Collapse
|
25
|
Tiwari P, Shukla RP, Yadav K, Singh N, Marwaha D, Gautam S, Bakshi AK, Rai N, Kumar A, Sharma D, Mishra PR. Dacarbazine-primed carbon quantum dots coated with breast cancer cell-derived exosomes for improved breast cancer therapy. J Control Release 2024; 365:43-59. [PMID: 37935257 DOI: 10.1016/j.jconrel.2023.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Imprecise targeting of chemotherapeutic drugs often leads to severe toxicity during breast cancer therapy. To address this issue, we have devised a strategy to load dacarbazine (DC) into fucose-based carbon quantum dots (CQDs), which are subsequently coated with exosomes (Ex-DC@CQDs) derived from breast cancer cells. Nanoparticle tracking analysis and western blotting revealed that Ex-DC@CQDs retained the structural and functional characteristics of exosomes. We found that exosomes facilitated the transport of DC@CQDs to cancer cells via heparan sulfate proteoglycan (HSPG) receptors, followed by an augmented depolarization of the mitochondrial membrane potential, ROS generation, and induction of apoptosis leading to cell death. In vivo imaging and pharmacokinetic studies demonstrated enhanced antitumor targeting and efficacy compared to free DC which we attribute to an improved pharmacokinetic profile, a greater tumor accumulation via exosome-mediated- HSPG receptor-driven cell uptake, and sustained release of the Ex-DC@CQDs. Our findings may pave the way for the further development of biologically sourced nanocarriers for breast cancer targeting.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Jawaharlal Nehru University, New Delhi, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ankit Kumar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Deepak Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
26
|
Sokol MB, Sokhraneva VA, Groza NV, Mollaeva MR, Yabbarov NG, Chirkina MV, Trufanova AA, Popenko VI, Nikolskaya ED. Thymol-Modified Oleic and Linoleic Acids Encapsulated in Polymeric Nanoparticles: Enhanced Bioactivity, Stability, and Biomedical Potential. Polymers (Basel) 2023; 16:72. [PMID: 38201737 PMCID: PMC10781094 DOI: 10.3390/polym16010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Unsaturated fatty acids, such as oleic acid (OA) and linoleic acid (LA), are promising antimicrobial and cytostatic agents. We modified OA and LA with thymol (TOA and TLA, respectively) to expand their bioavailability, stability, and possible applications, and encapsulated these derivatives in polymeric nanoparticles (TOA-NPs and TLA-NPs, respectively). Prior to synthesis, we performed mathematical simulations with PASS and ADMETlab 2.0 to predict the biological activity and pharmacokinetics of TOA and TLA. TOA and TLA were synthesized via esterification in the presence of catalysts. Next, we formulated nanoparticles using the single-emulsion solvent evaporation technique. We applied dynamic light scattering, Uv-vis spectroscopy, release studies under gastrointestinal (pH 1.2-6.8) and blood environment simulation conditions (pH 7.4), and in vitro biological activity testing to characterize the nanoparticles. PASS revealed that TOA and TLA have antimicrobial and anticancer therapeutic potential. ADMETlab 2.0 provided a rationale for TOA and TLA encapsulation. The nanoparticles had an average size of 212-227 nm, with a high encapsulation efficiency (71-93%), and released TOA and TLA in a gradual and prolonged mode. TLA-NPs possessed higher antibacterial activity against B. cereus and S. aureus and pronounced cytotoxic activity against MCF-7, K562, and A549 cell lines compared to TOA-NPs. Our findings expand the biomedical application of fatty acids and provide a basis for further in vivo evaluation of designed derivatives and formulations.
Collapse
Affiliation(s)
- Maria B. Sokol
- N.M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (M.R.M.); (N.G.Y.); (M.V.C.); (A.A.T.)
| | - Vera A. Sokhraneva
- N.A. Preobrazhensky Department of Chemistry and Technology of Biologically Active Compounds, Medicinal and Organic Chemistry, M.V. Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119571 Moscow, Russia; (V.A.S.); (N.V.G.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 11999 Moscow, Russia;
| | - Nataliya V. Groza
- N.A. Preobrazhensky Department of Chemistry and Technology of Biologically Active Compounds, Medicinal and Organic Chemistry, M.V. Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119571 Moscow, Russia; (V.A.S.); (N.V.G.)
| | - Mariia R. Mollaeva
- N.M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (M.R.M.); (N.G.Y.); (M.V.C.); (A.A.T.)
| | - Nikita G. Yabbarov
- N.M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (M.R.M.); (N.G.Y.); (M.V.C.); (A.A.T.)
| | - Margarita V. Chirkina
- N.M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (M.R.M.); (N.G.Y.); (M.V.C.); (A.A.T.)
| | - Anna A. Trufanova
- N.M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (M.R.M.); (N.G.Y.); (M.V.C.); (A.A.T.)
| | - Vladimir I. Popenko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 11999 Moscow, Russia;
| | - Elena D. Nikolskaya
- N.M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (M.R.M.); (N.G.Y.); (M.V.C.); (A.A.T.)
| |
Collapse
|
27
|
Yang C, Cao X, He L, Wu C, Zhao M, Duan F, Qiu Z, Zhu X, Yan Y, Li S, Li W, Shen B. Promoting Intratumoral Drug Accumulation by Bio-Membrane Regulated Active Targeting for Tumor Photothermal Therapy. Int J Nanomedicine 2023; 18:7287-7304. [PMID: 38076730 PMCID: PMC10710258 DOI: 10.2147/ijn.s434645] [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: 08/09/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Insufficient tumor permeability and inadequate nanoparticle retention continue to be significant limitations in the efficacy of anti-tumor drug therapy. Numerous studies have focused on enhancing tumor perfusion by improvement of tumor-induced endothelial leakage, often known as the enhanced permeability and retention (EPR) effect. However, these approaches have produced suboptimal therapeutic outcomes and have been associated with significant side effects. Therefore, in this study, we prepared tumor cell membrane-coated gold nanorods (GNR@TM) to enhance drug delivery in tumors through homogeneous targeting of tumor cell membranes and in situ real-time photo-controlled therapy. Methods Here, we fabricated GNR@TM, and characterized it using various techniques including Ultraviolet-Visible (UV-Vis) spectrophotometer, particle size analysis, potential measurement, and transmission electron microscopy (TEM). The cellular uptake and cytotoxicity of GNR@TM were analyzed by flow cytometry, confocal laser scanning microscopy (CLSM), TEM, CCK8 assay and live/dead staining. Tissue drug distribution was determined by inductively coupled plasma mass spectrometry (ICP-MS) and immunofluorescence staining. Furthermore, to evaluate the therapeutic effect, mice bearing MB49 tumors were intravenously administered with GNR@TM. Subsequently, near-infrared (NIR) laser therapy was performed, and the mice's tumor growth and body weight were monitored. Results The tumor cell membrane coating endowed GNR@TM with extended circulation time in vivo and homotypic targeting to tumor, thereby enhancing the accumulation of GNR@TM within tumors. Upon 780 nm laser, GNR@TM exhibited excellent photothermal conversion capability, leading to increased tumor vascular leakage. This magnification of the EPR effect induced by NIR laser further increased the accumulation of GNR@TM at the tumor site, demonstrating strong antitumor effects in vivo. Conclusion In this study, we successfully developed a NIR-triggered nanomedicine that increased drug accumulation in tumor through photo-controlled therapy and homotypic targeting of the tumor cell membrane. GNR@TM has been demonstrated effective suppression of tumor growth, excellent biocompatibility, and significant potential for clinical applications.
Collapse
Affiliation(s)
- Chenkai Yang
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiangqian Cao
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Lei He
- Department of Nanomedicine & Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai, People’s Republic of China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
| | - Cong Wu
- Department of Nanomedicine & Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai, People’s Republic of China
| | - Mengxin Zhao
- Department of Nanomedicine & Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai, People’s Republic of China
| | - Fei Duan
- Department of Nanomedicine & Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai, People’s Republic of China
| | - Zhiwen Qiu
- Department of Nanomedicine & Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai, People’s Republic of China
| | - Xiaodong Zhu
- Department of Nanomedicine & Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai, People’s Republic of China
| | - Yilin Yan
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Shengzhou Li
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Wei Li
- Department of Nanomedicine & Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai, People’s Republic of China
| | - Bing Shen
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
28
|
Zhang X, Pan J, Ye X, Chen Y, Wang L, Meng X, Chen W, Wang F. Activation of CYP3A by Accelerated Blood Clearance Phenomenon Potentiates the Hepatocellular Carcinoma-Targeting Therapeutic Effects of PEGylated Anticancer Prodrug Liposomes. Drug Metab Dispos 2023; 51:1651-1662. [PMID: 37775330 DOI: 10.1124/dmd.123.001496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/14/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023] Open
Abstract
Reduced enzyme activity in hepatocellular carcinoma (HCC) and poor targeting limit the application of enzyme-activating prodrugs, which is also detrimental to the effective treatment of HCC. Here, we investigated whether accelerated blood clearance (ABC) phenomenon occurs in HCC models following repeated injections of PEGylated liposomes (PEG-L), thus inducing prodrug accumulation and activation in the liver and exerting highly effective and low-toxicity therapeutic effects on HCC. First, PEGylated liposomal cyclophosphamide was prepared by solvent injection and characterized. Importantly, preinjection of PEG-L induced the ABC phenomenon and activation of CYP3A in both HCC rats and HCC mice by studying the effects of repeated injections of PEG-L on pharmacokinetics and tissue distribution. Next, the efficacy and toxicity of repeated injections of PEG-L in HCC mice were examined, and our data indicate that repeated injections are administered in a manner that significantly enhances the antitumor effect compared with controls, with little or no toxicity to other organs. To further reveal the pharmacokinetic mechanism of PEG-L repeated administration for the treatment of HCC, the protein expression of hepatic CYP3A and the concentration of cyclophosphamide in the liver and spleen of HCC mice by inhibiting CYP3A were analyzed. These results revealed that inducing CYP3A to accelerate the rapid conversion of prodrugs that accumulate significantly in the liver is a key mechanism for the treatment of HCC with repeated injections of PEG-L. Collectively, this work taps into the application potential of the ABC phenomenon and provides new insights into the clinical application of PEGylated nanoformulations. SIGNIFICANCE STATEMENT: This study revealed that repeated injections of PEGylated liposomes could induce the accelerated blood clearance (ABC) phenomenon characterized by hepatic accumulation and CYP3A activation based on hepatocellular carcinoma (HCC) rats and HCC mice. Furthermore, it was verified that induction of the ABC phenomenon dependent on hepatic accumulation and CYP3A activation could enhance the antihepatocellular carcinoma effects of PEGylated anticancer prodrugs in HCC mice. This elucidated the relevant pharmacokinetic mechanisms and unearthed new clues for solving the clinical application of PEGylated nanoparticles.
Collapse
Affiliation(s)
- Xue Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China (X.Z., J.P., L.W., W.C., F.W.); Hefei Hospital Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, China (X.Y., X.M., F.W.); School of Pharmacy, Anhui Medical University, Hefei, China (F.W.); The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, China (F.W.); and Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China (Y.C.)
| | - Jianquan Pan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China (X.Z., J.P., L.W., W.C., F.W.); Hefei Hospital Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, China (X.Y., X.M., F.W.); School of Pharmacy, Anhui Medical University, Hefei, China (F.W.); The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, China (F.W.); and Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China (Y.C.)
| | - Xi Ye
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China (X.Z., J.P., L.W., W.C., F.W.); Hefei Hospital Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, China (X.Y., X.M., F.W.); School of Pharmacy, Anhui Medical University, Hefei, China (F.W.); The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, China (F.W.); and Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China (Y.C.)
| | - Yunna Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China (X.Z., J.P., L.W., W.C., F.W.); Hefei Hospital Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, China (X.Y., X.M., F.W.); School of Pharmacy, Anhui Medical University, Hefei, China (F.W.); The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, China (F.W.); and Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China (Y.C.)
| | - Lei Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China (X.Z., J.P., L.W., W.C., F.W.); Hefei Hospital Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, China (X.Y., X.M., F.W.); School of Pharmacy, Anhui Medical University, Hefei, China (F.W.); The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, China (F.W.); and Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China (Y.C.)
| | - Xiangyun Meng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China (X.Z., J.P., L.W., W.C., F.W.); Hefei Hospital Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, China (X.Y., X.M., F.W.); School of Pharmacy, Anhui Medical University, Hefei, China (F.W.); The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, China (F.W.); and Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China (Y.C.)
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China (X.Z., J.P., L.W., W.C., F.W.); Hefei Hospital Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, China (X.Y., X.M., F.W.); School of Pharmacy, Anhui Medical University, Hefei, China (F.W.); The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, China (F.W.); and Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China (Y.C.).
| | - Fengling Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China (X.Z., J.P., L.W., W.C., F.W.); Hefei Hospital Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, China (X.Y., X.M., F.W.); School of Pharmacy, Anhui Medical University, Hefei, China (F.W.); The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, China (F.W.); and Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China (Y.C.).
| |
Collapse
|
29
|
Cooley MB, Wegierak D, Perera R, Abenojar EC, Nittayacharn PA, Berg FM, Kim Y, Kolios MC, Exner AA. Assessing Tumor Microenvironment Characteristics and Stratifying EPR with a Nanobubble Companion Nanoparticle via Contrast-Enhanced Ultrasound Imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567934. [PMID: 38045236 PMCID: PMC10690218 DOI: 10.1101/2023.11.20.567934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
The tumor microenvironment is characterized by dysfunctional endothelial cells, resulting in heightened vascular permeability. Many nanoparticle-based drug delivery systems attempt to use this enhanced permeability combined with impaired lymphatic drainage (a concept known as the 'enhanced permeability and retention effect' or EPR effect) as the primary strategy for drug delivery, but this has not proven to be as clinically effective as anticipated. The specific mechanisms behind the inconsistent clinical outcomes of nanotherapeutics have not been clearly articulated, and the field has been hampered by a lack of accessible tools to study EPR-associated phenomena in clinically relevant scenarios. While medical imaging has tremendous potential to contribute to this area, it has not been broadly explored. This work examines, for the first time, the use of multiparametric dynamic contrast-enhanced ultrasound (CEUS) with a novel nanoscale contrast agent to examine tumor microenvironment characteristics noninvasively and in real-time. We demonstrate that CEUS imaging can: (1) evaluate tumor microenvironment features and (2) be used to help predict the distribution of doxorubicin-loaded liposomes in the tumor parenchyma. CEUS using nanobubbles (NBs) was carried out in two tumor types of high (LS174T) and low (U87) vascular permeability, and time-intensity curve (TIC) parameters were evaluated in both models prior to injection of doxorubicin liposomes. Consistently, LS174T tumors showed significantly different TIC parameters, including area under the rising curve (2.7x), time to peak intensity (1.9x) and decorrelation time (DT, 1.9x) compared to U87 tumors. Importantly, the DT parameter successfully predicted tumoral nanoparticle distribution (r = 0.86 ± 0.13). Ultimately, substantial differences in NB-CEUS generated parameters between LS174T and U87 tumors suggest that this method may be useful in determining tumor vascular permeability and could be used as a biomarker for identifying tumor characteristics and predicting sensitivity to nanoparticle-based therapies. These findings could ultimately be applied to predicting treatment efficacy and to evaluating EPR in other diseases with pathologically permeable vasculature.
Collapse
|
30
|
Khan S, Falahati M, Cho WC, Vahdani Y, Siddique R, Sharifi M, Jaragh-Alhadad LA, Haghighat S, Zhang X, Ten Hagen TLM, Bai Q. Core-shell inorganic NP@MOF nanostructures for targeted drug delivery and multimodal imaging-guided combination tumor treatment. Adv Colloid Interface Sci 2023; 321:103007. [PMID: 37812992 DOI: 10.1016/j.cis.2023.103007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 08/16/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
It is well known that metal-organic framework (MOF) nanostructures have unique characteristics such as high porosity, large surface areas and adjustable functionalities, so they are ideal candidates for developing drug delivery systems (DDSs) as well as theranostic platforms in cancer treatment. Despite the large number of MOF nanostructures that have been discovered, conventional MOF-derived nanosystems only have a single biofunctional MOF source with poor colloidal stability. Accordingly, developing core-shell MOF nanostructures with good colloidal stability is a useful method for generating efficient drug delivery, multimodal imaging and synergistic therapeutic systems. The preparation of core-shell MOF nanostructures has been done with a variety of materials, but inorganic nanoparticles (NPs) are highly effective for drug delivery and imaging-guided tumor treatment. Herein, we aimed to overview the synthesis of core-shell inorganic NP@MOF nanostructures followed by the application of core-shell MOFs derived from magnetic, quantum dots (QDs), gold (Au), and gadolinium (Gd) NPs in drug delivery and imaging-guided tumor treatment. Afterward, we surveyed different factors affecting prolonged drug delivery and cancer therapy, cellular uptake, biocompatibility, biodegradability, and enhanced permeation and retention (EPR) effect of core-shell MOFs. Last but not least, we discussed the challenges and the prospects of the field. We envision this article may hold great promise in providing valuable insights regarding the application of hybrid nanostructures as promising and potential candidates for multimodal imaging-guided combination cancer therapy.
Collapse
Affiliation(s)
- Suliman Khan
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mojtaba Falahati
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus MC, Rotterdam, the Netherlands.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China
| | - Yasaman Vahdani
- Department of Biochemistry and Molecular Medicine, University of Montreal, Canada
| | - Rabeea Siddique
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran; Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | | | - Setareh Haghighat
- Department of Microbiology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus MC, Rotterdam, the Netherlands.
| | - Qian Bai
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
31
|
Agrawal A, Leng Q, Imtiyaz Z, Mixson AJ. Exploring the outer limits of polyplexes. Biochem Biophys Res Commun 2023; 678:33-38. [PMID: 37619309 PMCID: PMC10528873 DOI: 10.1016/j.bbrc.2023.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023]
Abstract
Histidine-containing polymers show promise in their transport of nucleic acids in vitro and in vivo. In addition to the pH-buffering histidine component, the polymer often contains a protonated component at physiological pH, such as lysine. These polyplexes usually accumulate in the tumor by enhanced permeability and retention, which has proved disappointing in clinical trials. We presently compare two histidine-lysine (HK) peptide polyplexes for their neuropilin-1-mediated transport of plasmids in vivo. While the polymerized HK (H2KC-48) polyplex was markedly better than the monomeric HK (H2K) polyplex in vitro, both HK polyplexes were effective in transfecting tumor xenografts over a wide range of peptide and plasmid ratios. Nevertheless, polyplexes of low peptide/DNA ratios gave higher tumor transfection and specificity than those of higher ratios. Surprisingly, there was minimal to no gel retardation of polyplexes made from these low ratios during electrophoresis. These results demonstrate that loosely packed HK polyplexes effectively transfected tumors in vivo.
Collapse
Affiliation(s)
- A Agrawal
- Department of Pathology, University of Maryland School of Medicine, 10 S. Pine St., University of Maryland, Baltimore, MD, 21201, USA
| | - Q Leng
- Department of Pathology, University of Maryland School of Medicine, 10 S. Pine St., University of Maryland, Baltimore, MD, 21201, USA
| | - Z Imtiyaz
- Department of Pathology, University of Maryland School of Medicine, 10 S. Pine St., University of Maryland, Baltimore, MD, 21201, USA
| | - A James Mixson
- Department of Pathology, University of Maryland School of Medicine, 10 S. Pine St., University of Maryland, Baltimore, MD, 21201, USA.
| |
Collapse
|
32
|
Wu Q, Hu Y, Yu B, Hu H, Xu FJ. Polysaccharide-based tumor microenvironment-responsive drug delivery systems for cancer therapy. J Control Release 2023; 362:19-43. [PMID: 37579973 DOI: 10.1016/j.jconrel.2023.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/05/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
The biochemical indicators of tumor microenvironment (TME) that are different from normal tissues provide the possibility for constructing intelligent drug delivery systems (DDSs). Polysaccharides with good biocompatibility, biodegradability, and unique biological properties are ideal materials for constructing DDSs. Nanogels, micelles, organic-inorganic nanocomposites, hydrogels, and microneedles (MNs) are common polysaccharide-based DDSs. Polysaccharide-based DDSs enable precise control of drug delivery and release processes by incorporating TME-specific biochemical indicators. The classification and design strategies of polysaccharide-based TME-responsive DDSs are comprehensively reviewed. The advantages and challenges of current polysaccharide-based DDSs are summarized and the future directions of development are foreseen. The polysaccharide-based TME-responsive DDSs are expected to provide new strategies and solutions for cancer therapy and make important contributions to the realization of precision medicine.
Collapse
Affiliation(s)
- Qimeng Wu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Yang Hu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Hao Hu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China.
| | - Fu-Jian Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
33
|
Wang P, Yang Y, Wen H, Li D, Zhang H, Wang Y. Progress in construction and release of natural polysaccharide-platinum nanomedicines: A review. Int J Biol Macromol 2023; 250:126143. [PMID: 37544564 DOI: 10.1016/j.ijbiomac.2023.126143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Natural polysaccharides are natural biomaterials that have become candidate materials for nano-drug delivery systems due to their excellent biodegradability and biocompatibility. Platinum (Pt) drugs have been widely used in the clinical therapy for various solid tumors. However, their extensive systemic toxicity and the drug resistance acquired by cancer cells limit the applications of platinum drugs. Modern nanobiotechnology provides the possibility for targeted delivery of platinum drugs to the tumor site, thereby minimizing toxicity and optimizing the efficacies of the drugs. In recent years, numerous natural polysaccharide-platinum nanomedicine delivery carriers have been developed, such as nanomicelles, nanospheres, nanogels, etc. Herein, we provide an overview on the construction and drug release of natural polysaccharide-Pt nanomedicines in recent years. Current challenges and future prospectives in this field are also put forward. In general, combining with irradiation and tumor microenvironment provides a significant research direction for the construction of natural polysaccharide-platinum nanomedicines and the release of responsive drugs in the future.
Collapse
Affiliation(s)
- Pengge Wang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China; College of Biological and Pharmaceutical Engineering, Nanjing University of Technology, Nanjing City, Jiangsu Province 211816, China
| | - Yunxia Yang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China; Jiangsu Province Engineering Research Center of Agricultural Breeding Pollution Control and Resource, Yancheng Teachers University, Yancheng 224007, China; Jiangsu Key Laboratory for Bioresources of Saline Soils, Yancheng Teachers University, Yancheng 224007, China.
| | - Haoyu Wen
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Dongqing Li
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Hongmei Zhang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Yanqing Wang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China.
| |
Collapse
|
34
|
Ding N, Luo G, Li H, Xing C, Gao Y, Xi W, Wu W, Wang D, Zheng L, Kang Y, Chi X. A Cyclodextrin-Based pH-Responsive MicroRNA Delivery Platform Targeting Polarization of M1 to M2 Macrophages for Sepsis Therapy. Adv Healthc Mater 2023; 12:e2301243. [PMID: 37463303 DOI: 10.1002/adhm.202301243] [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: 04/20/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/20/2023]
Abstract
The mortality rate of sepsis remains high despite improvements in the diagnosis and treatment of sepsis using symptomatic and supportive therapies, such as anti-infection therapy and fluid resuscitation. Nucleic acid-based drugs have therapeutic potential, although their poor stability and low delivery efficiency have hindered their widespread use. Herein, it is confirmed that miR-223 can polarize proinflammation M1 macrophages to anti-inflammation M2 macrophages. A pH-sensitive nano-drug delivery system comprising β-cyclodextrin-poly(2-(diisopropylamino)ethyl methacrylate)/distearoyl phosphoethanolamine-polyethylene glycol (β-CD-PDPA/DSPE-PEG) is synthesized and developed to target M1 macrophages and miR-223 is encapsulated into nanoparticles (NPs) for sepsis treatment. NPs/miR-223 demonstrated in vitro pH responsiveness with favorable biosafety, stability, and high delivery efficiency. In vivo studies demonstrate that NPs/miR-223 are preferentially accumulated and retained in the inflammation site, thereby reducing inflammation and improving the survival rate of mice with sepsis while exhibiting ideal biosafety. Mechanically, NPs/miR-223 regulates macrophage polarization by targeting Pknox1 and inhibiting the activation of the NF-κB signaling pathway, thereby achieving an anti-inflammatory effect. Collectively, it is demonstrated that the miRNA delivery vector described here provides a new approach for sepsis treatment and accelerates the advancement of nucleic acid drug therapy.
Collapse
Affiliation(s)
- Ni Ding
- Department of Anaesthesiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Gangjian Luo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Huiting Li
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Chengyuan Xing
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Yuanji Gao
- College of Chemistry and Materials Science, Sichuan Normal University, Chengdu, 610068, China
| | - Wenjie Xi
- Department of Anaesthesiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Weijie Wu
- Department of Anaesthesiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Dan Wang
- Department of Anaesthesiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Lei Zheng
- Department of Anaesthesiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Yang Kang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinjin Chi
- Department of Anaesthesiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
35
|
Oh H, Jeong E, Lee JS, Kim J, Lee D, Kim BS, Sung D, Koo H, Choi WI, Tae G. ROS-responsive PEGylated ferrocene polymer nanoparticles with improved stability for tumor-selective chemotherapy and imaging. Mater Today Bio 2023; 22:100774. [PMID: 37664795 PMCID: PMC10468360 DOI: 10.1016/j.mtbio.2023.100774] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/27/2023] [Accepted: 08/15/2023] [Indexed: 09/05/2023] Open
Abstract
Ferrocene-based nanoparticles have garnered interest as reactive oxygen species (ROS)-responsive nanocarriers of anticancer drugs and imaging agents. However, their biomedical applications remain limited due to their poor physiological stability. PEGylation of nanocarriers improves their stability and biocompatibility. In this study, we aimed to develop novel PEG-ferrocene nanoparticles (PFNPs) with enhanced stability and ROS responsiveness for the delivery of paclitaxel (PTX) and imaging agents. PEGylation improved the stability of ferrocene nanoparticles, inhibiting their ROS-responsive destruction. Several PEG-ferrocene polymers containing different molar ratios of methacrylic acid and poly (ethylene glycol) methyl ether methacrylate was designed for optimization. ROS-responsive polymers with optimal monomer ratios were self-assembled into PFNPs with enhanced stability. The PFNPs distended, effectively releasing encapsulated PTX and imaging agents within 8 h in the presence of ROS. Furthermore, they remained stable, with no changes in their hydrodynamic diameters or polydispersity indexes after storage in an aqueous solution and biological buffer. The accumulation of PFNPs in a tumor model in vivo was 15-fold higher than a free dye. PTX-loaded PFNPs showed a substantial tumor-suppression effect, reducing tumor size to approximately 18% of that in the corresponding control group. These findings suggest a promising application of ROS-responsive PFNPs in tumor treatment as biocompatible nanocarriers of anticancer drugs and imaging agents.
Collapse
Affiliation(s)
- Hyeryeon Oh
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, 28160, Republic of Korea
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Eunjin Jeong
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Jin Sil Lee
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, 28160, Republic of Korea
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jisu Kim
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, 28160, Republic of Korea
| | - Donghyun Lee
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Byoung Soo Kim
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, 28160, Republic of Korea
| | - Daekyung Sung
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, 28160, Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Won Il Choi
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, 28160, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| |
Collapse
|
36
|
AbdEl-haq M, Kumar A, Ait Mohand FE, Kravchenko-Balasha N, Rottenberg Y, Domb AJ. Paclitaxel Delivery to the Brain for Glioblastoma Treatment. Int J Mol Sci 2023; 24:11722. [PMID: 37511480 PMCID: PMC10380674 DOI: 10.3390/ijms241411722] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
The development of paclitaxel-loaded polymeric nanoparticles for the treatment of brain tumors was investigated. Poly(lactide-glycolide) (PLGA) nanoparticles containing 10% w/w paclitaxel with a particle size of 216 nm were administered through intranasal and intravenous routes to male Sprague-Dawley rats at a dose of 5 mg/kg. Both routes of administration showed appreciable accumulation of paclitaxel in brain tissue, liver, and kidney without any sign of toxicity. The anti-proliferative effect of the nanoparticles on glioblastoma tumor cells was comparable to that of free paclitaxel.
Collapse
Affiliation(s)
- Muhammad AbdEl-haq
- Institute of Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Awanish Kumar
- Institute of Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Fatima-ezzahra Ait Mohand
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel (N.K.-B.)
| | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel (N.K.-B.)
| | - Yakir Rottenberg
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel;
| | - Abraham J. Domb
- Institute of Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| |
Collapse
|
37
|
Shabatina TI, Vernaya OI, Shimanovskiy NL, Melnikov MY. Metal and Metal Oxides Nanoparticles and Nanosystems in Anticancer and Antiviral Theragnostic Agents. Pharmaceutics 2023; 15:pharmaceutics15041181. [PMID: 37111666 PMCID: PMC10141702 DOI: 10.3390/pharmaceutics15041181] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
The development of antiviral treatment and anticancer theragnostic agents in recent decades has been associated with nanotechnologies, and primarily with inorganic nanoparticles (INPs) of metal and metal oxides. The large specific surface area and its high activity make it easy to functionalize INPs with various coatings (to increase their stability and reduce toxicity), specific agents (allowing retention of INPs in the affected organ or tissue), and drug molecules (for antitumor and antiviral therapy). The ability of magnetic nanoparticles (MNPs) of iron oxides and ferrites to enhance proton relaxation in specific tissues and serve as magnetic resonance imaging contrast agents is one of the most promising applications of nanomedicine. Activation of MNPs during hyperthermia by an external alternating magnetic field is a promising method for targeted cancer therapy. As therapeutic tools, INPs are promising carriers for targeted delivery of pharmaceuticals (either anticancer or antiviral) via magnetic drug targeting (in case of MNPs), passive or active (by attaching high affinity ligands) targeting. The plasmonic properties of Au nanoparticles (NPs) and their application for plasmonic photothermal and photodynamic therapies have been extensively explored recently in tumor treatment. The Ag NPs alone and in combination with antiviral medicines reveal new possibilities in antiviral therapy. The prospects and possibilities of INPs in relation to magnetic hyperthermia, plasmonic photothermal and photodynamic therapies, magnetic resonance imaging, targeted delivery in the framework of antitumor theragnostic and antiviral therapy are presented in this review.
Collapse
Affiliation(s)
- Tatyana I Shabatina
- Department of Chemistry, M.V. Lomonosov Moscow State University, Leninskie Gori Build. 1/3, Moscow 119991, Russia
- Faculty of Fundamental Sciences, N.E. Bauman Moscow Technical University, Moscow 105005, Russia
| | - Olga I Vernaya
- Department of Chemistry, M.V. Lomonosov Moscow State University, Leninskie Gori Build. 1/3, Moscow 119991, Russia
- Faculty of Fundamental Sciences, N.E. Bauman Moscow Technical University, Moscow 105005, Russia
| | - Nikolay L Shimanovskiy
- Department of Molecular Pharmacology and Radiobiology, N.I. Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Mikhail Ya Melnikov
- Department of Chemistry, M.V. Lomonosov Moscow State University, Leninskie Gori Build. 1/3, Moscow 119991, Russia
| |
Collapse
|
38
|
State-of-the-art advancement of surface functionalized layered double hydroxides for cell-specific targeting of therapeutics. Adv Colloid Interface Sci 2023; 314:102869. [PMID: 36933542 DOI: 10.1016/j.cis.2023.102869] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/14/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Over the years, layered double hydroxides (LDHs) hold a specific position in biomedicine due to their tunable chemical composition and appropriate structural properties. However, LDHs lack adequate sensitivity for active targeting because of less active surface area and low mechanical strength in physiological conditions. The exploitation of eco-friendly materials, such as chitosan (CS), for surface engineering of LDHs, whose payloads are transferred only under certain conditions, can help develop stimuli-responsive materials owing to high biosafety and unique mechanical strength. We aim to render a well-oriented scenario toward the latest achievements of a bottom-up technology relying on the surface functionalization of LDHs to fabricate functional formulations with promoted bio-functionality and high encapsulation efficiency for various bioactives. Many efforts have been devoted to critical aspects of LDHs, including systemic biosafety and the suitability for developing multicomponent systems via integration with therapeutic modalities, which are thoroughly discussed herein. In addition, a comprehensive discussion was provided for the recent progress in the emergence of CS-coated LDHs. Finally, the challenges and future perspectives in the fabrication of efficient CS-LDHs in biomedicine are considered, with a special focus on cancer treatment.
Collapse
|
39
|
Subhan MA, Parveen F, Filipczak N, Yalamarty SSK, Torchilin VP. Approaches to Improve EPR-Based Drug Delivery for Cancer Therapy and Diagnosis. J Pers Med 2023; 13:jpm13030389. [PMID: 36983571 PMCID: PMC10051487 DOI: 10.3390/jpm13030389] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
The innovative development of nanomedicine has promised effective treatment options compared to the standard therapeutics for cancer therapy. However, the efficiency of EPR-targeted nanodrugs is not always pleasing as it is strongly prejudiced by the heterogeneity of the enhanced permeability and retention effect (EPR). Targeting the dynamics of the EPR effect and improvement of the therapeutic effects of nanotherapeutics by using EPR enhancers is a vital approach to developing cancer therapy. Inadequate data on the efficacy of EPR in humans hampers the clinical translation of cancer drugs. Molecular targeting, physical amendment, or physiological renovation of the tumor microenvironment (TME) are crucial approaches for improving the EPR effect. Advanced imaging technologies for the visualization of EPR-induced nanomedicine distribution in tumors, and the use of better animal models, are necessary to enhance the EPR effect. This review discusses strategies to enhance EPR effect-based drug delivery approaches for cancer therapy and imaging technologies for the diagnosis of EPR effects. The effort of studying the EPR effect is beneficial, as some of the advanced nanomedicine-based EPR-enhancing approaches are currently undergoing clinical trials, which may be helpful to improve EPR-induced drug delivery and translation to clinics.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
- Correspondence: (M.A.S.); (V.P.T.)
| | - Farzana Parveen
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Punjab 63100, Pakistan
- Department of Pharmacy Services, DHQ Hospital Jhang 35200, Primary and Secondary Healthcare Department, Government of Punjab, Lahore, Punjab 54000, Pakistan
| | - Nina Filipczak
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | | | - Vladimir P. Torchilin
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
- Correspondence: (M.A.S.); (V.P.T.)
| |
Collapse
|
40
|
Wang F, Li N, Wang W, Ma L, Sun Y, Wang H, Zhan J, Yu D. A Multifunctional, Highly Biocompatible, and Double-Triggering Caramelized Nanotheranostic System Loaded with Fe 3O 4 and DOX for Combined Chemo-Photothermal Therapy and Real-Time Magnetic Resonance Imaging Monitoring of Triple Negative Breast Cancer. Int J Nanomedicine 2023; 18:881-897. [PMID: 36844435 PMCID: PMC9948638 DOI: 10.2147/ijn.s393507] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/14/2023] [Indexed: 02/20/2023] Open
Abstract
Purpose Owing to lack of specific molecular targets, the current clinical therapeutic strategy for triple negative breast cancer (TNBC) is still limited. In recent years, some nanosystems for malignancy treatment have received considerable attention. In this study, we prepared caramelized nanospheres (CNSs) loaded with doxorubicin (DOX) and Fe3O4 to achieve the synergistic effect of combined therapy and real-time magnetic resonance imaging (MRI) monitoring, so as to improve the diagnosis and therapeutic effect of TNBC. Methods CNSs with biocompatibility and unique optical properties were prepared by hydrothermal method, DOX and Fe3O4 were loaded on it to obtain Fe3O4/DOX@CNSs nanosystem. Characteristics including morphology, hydrodynamic size, zeta potentials and magnetic properties of Fe3O4/DOX@CNSs were evaluated. The DOX release was evaluated by different pH/near-infrared (NIR) light energy. Biosafety, pharmacokinetics, MRI and therapeutic treatment of Fe3O4@CNSs, DOX and Fe3O4/DOX@CNSs were examined in vitro or in vivo. Results Fe3O4/DOX@CNSs has an average particle size of 160 nm and a zeta potential of 27.5mV, it demonstrated that Fe3O4/DOX@CNSs is a stable and homogeneous dispersed system. The hemolysis experiment of Fe3O4/DOX@CNSs proved that it can be used in vivo. Fe3O4/DOX@CNSs displayed high photothermal conversion efficiency, extensive pH/heat-induced DOX release. 70.3% DOX release is observed under the 808 nm laser in the pH = 5 PBS solution, obviously higher than pH = 5 (50.9%) and pH = 7.4 (less than 10%). Pharmacokinetic experiments indicated the t1/2β, and AUC0-t of Fe3O4/DOX@CNSs were 1.96 and 1.31 -fold higher than those of DOX solution, respectively. Additionally, Fe3O4/DOX@CNSs with NIR had the greatest tumor suppression in vitro and in vivo. Moreover, this nanosystem demonstrated distinct contrast enhancement on T2 MRI to achieve real-time imaging monitoring during treatment. Conclusion Fe3O4/DOX@CNSs is a highly biocompatible, double-triggering and improved DOX bioavailability nanosystem that combines chemo-PTT and real-time MRI monitoring to achieve integration of diagnosis and treatment of TNBC.
Collapse
Affiliation(s)
- Fangqing Wang
- Department of Radiology, Qilu Hospital, Shandong University, Affiliated Hospital of Shandong University, Jinan, 250012, People’s Republic of China
| | - Nianlu Li
- Physical and Chemical Laboratory, Shandong Academy of Occupational Health and Occupational Medicine, Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250002, People’s Republic of China
| | - Wenbo Wang
- Department of Radiology, Qilu Hospital, Shandong University, Affiliated Hospital of Shandong University, Jinan, 250012, People’s Republic of China
| | - Long Ma
- The Testing Center of Shandong Bureau of China Metallurgical Geology Bureau, Shandong Normal University, Jinan, 250014, People’s Republic of China
| | - Yaru Sun
- Department of Nuclear Medicine, The Second Hospital of Shandong University, Affiliated Hospital of Shandong University, Jinan, 250033, People’s Republic of China
| | - Hong Wang
- Department of Radiology, Qilu Hospital, Shandong University, Affiliated Hospital of Shandong University, Jinan, 250012, People’s Republic of China
| | - Jinhua Zhan
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, People’s Republic of China,Correspondence: Jinhua Zhan, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, People’s Republic of China, Email
| | - Dexin Yu
- Department of Radiology, Qilu Hospital, Shandong University, Affiliated Hospital of Shandong University, Jinan, 250012, People’s Republic of China,Dexin Yu, Department of Radiology, Qilu Hospital, Shandong University, Affiliated Hospital of Shandong University, Jinan, 250012, People’s Republic of China, Tel +86-18560081629, Fax +86-531-86927544, Email
| |
Collapse
|
41
|
Zhang C, Song Y, Yan G, Ma J. Fluorinated carboxymethyl chitosan-based nano-prodrugs for precisely synergistic chemotherapy. Int J Biol Macromol 2023; 227:252-261. [PMID: 36549609 DOI: 10.1016/j.ijbiomac.2022.12.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/04/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
The clinical transformation of polysaccharide-based nano-prodrugs remains a long way off, due to the shackles on easy metabolic clearance, dilemma of dose-dependent toxicity and immunogenicity, and poor tumor selectivity. To address these challenges, the fluorinated dual-crosslinked carboxymethyl chitosan (CMCS)-based nano-prodrugs with precise structure were facilely developed through the reaction of CMCS with water-soluble stimuli-responsive synergistic small molecule prodrug (Pt(IV)-1), glutaraldehyde and heptafluorobutyric anhydride successively. The fluorination enabled the nano-prodrugs to display metabolic stability and improve tumoral cellular uptake. The pH/glutathione (GSH)-sensitive dual-crosslinked structure enabled the nano-prodrugs to show physicochemical stability at physiological pH, selective drug release and synergistic cytotoxicity at tumoral intracellular pH/GSH, and circumventing the dilemma of dose-dependent toxicity and immunogenicity induced by that crosslinked or grafted via a single drug. These superior performances promoted stability in long-term storage and circulation, normal blood routine and aminotransferase, fantastic hemocompatibility, selective tumor accumulation and precisely synergistic chemotherapy, therefore achieving significant tumor growth inhibition while minimizing side effects. Thus, the precise fluorinated dual-crosslinked CMCS-based nano-prodrugs have great potential for selective clinical cancer treatment.
Collapse
Affiliation(s)
- Chensong Zhang
- Anhui Medical University, Hefei 230000, China; Department of Oncology Surgery, First Affiliated Hospital of Bengbu Medical College Bengbu, 233000, China
| | - Yining Song
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui 233030, China
| | - Guoqing Yan
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, China.
| | - Jiachi Ma
- Anhui Medical University, Hefei 230000, China; Department of Oncology Surgery, First Affiliated Hospital of Bengbu Medical College Bengbu, 233000, China.
| |
Collapse
|
42
|
Highlights of New Strategies to Increase the Efficacy of Transition Metal Complexes for Cancer Treatments. Molecules 2022; 28:molecules28010273. [PMID: 36615466 PMCID: PMC9822110 DOI: 10.3390/molecules28010273] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023] Open
Abstract
Although important progress has been made, cancer still remains a complex disease to treat. Serious side effects, the insurgence of resistance and poor selectivity are some of the problems associated with the classical metal-based anti-cancer therapies currently in clinical use. New treatment approaches are still needed to increase cancer patient survival without cancer recurrence. Herein, we reviewed two promising-at least in our opinion-new strategies to increase the efficacy of transition metal-based complexes. First, we considered the possibility of assembling two biologically active fragments containing different metal centres into the same molecule, thus obtaining a heterobimetallic complex. A critical comparison with the monometallic counterparts was done. The reviewed literature has been divided into two groups: the case of platinum; the case of gold. Secondly, the conjugation of metal-based complexes to a targeting moiety was discussed. Particularly, we highlighted some interesting examples of compounds targeting cancer cell organelles according to a third-order targeting approach, and complexes targeting the whole cancer cell, according to a second-order targeting strategy.
Collapse
|
43
|
Rodrigues CF, Fernandes N, de Melo‐Diogo D, Correia IJ, Moreira AF. Cell-Derived Vesicles for Nanoparticles' Coating: Biomimetic Approaches for Enhanced Blood Circulation and Cancer Therapy. Adv Healthc Mater 2022; 11:e2201214. [PMID: 36121767 PMCID: PMC11481079 DOI: 10.1002/adhm.202201214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/11/2022] [Indexed: 01/28/2023]
Abstract
Cancer nanomedicines are designed to encapsulate different therapeutic agents, prevent their premature release, and deliver them specifically to cancer cells, due to their ability to preferentially accumulate in tumor tissue. However, after intravenous administration, nanoparticles immediately interact with biological components that facilitate their recognition by the immune system, being rapidly removed from circulation. Reports show that less than 1% of the administered nanoparticles effectively reach the tumor site. This suboptimal pharmacokinetic profile is pointed out as one of the main factors for the nanoparticles' suboptimal therapeutic effectiveness and poor translation to the clinic. Therefore, an extended blood circulation time may be crucial to increase the nanoparticles' chances of being accumulated in the tumor and promote a site-specific delivery of therapeutic agents. For that purpose, the understanding of the forces that govern the nanoparticles' interaction with biological components and the impact of the physicochemical properties on the in vivo fate will allow the development of novel and more effective nanomedicines. Therefore, in this review, the nano-bio interactions are summarized. Moreover, the application of cell-derived vesicles for extending the blood circulation time and tumor accumulation is reviewed, focusing on the advantages and shortcomings of each cell source.
Collapse
Affiliation(s)
- Carolina F. Rodrigues
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Natanael Fernandes
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Duarte de Melo‐Diogo
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Ilídio J. Correia
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - André F. Moreira
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
- CPIRN‐UDI/IPG – Center of Potential and Innovation in Natural Resources, Research Unit for Inland DevelopmentInstituto Politécnico da GuardaAvenida Dr. Francisco de Sá CarneiroGuarda6300‐559Portugal
| |
Collapse
|
44
|
Nanostrategies: The Future Medicine for Fighting Cancer Progression and Drug Resistance 2.0. Int J Mol Sci 2022; 23:ijms232214486. [PMID: 36430963 PMCID: PMC9695259 DOI: 10.3390/ijms232214486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The attractiveness of the nanomaterials research field has persisted [...].
Collapse
|
45
|
Zou X, Li T, Mao Y, Zhu M, Chen X, Niu J, Dong T, Jiang J, Yang X. Multifunctional Drug-Loaded Phase-Change Nanoparticles Inhibit the Epithelial-Mesenchymal Transition of Hepatocellular Carcinoma by Affecting the Activity of Activated Hepatic Stellate Cells. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6441179. [PMID: 36411770 PMCID: PMC9675611 DOI: 10.1155/2022/6441179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 01/18/2024]
Abstract
Objectives. Preparation of a multifunctional drug-loaded phase-change nanoparticle (NP), pirfenidone perfluoropentane liposome NPs (PPL NPs), and combined with low-intensity focused ultrasound (LIFU) to influence epithelial mesenchymal transition (EMT) for hepatocellular carcinoma (HCC) by inhibiting the activity of activated Hepatic Stellate Cells (a-HSCs). Methods. PPL NPs were prepared by the thin film dispersion method. The appearance, particle size, zeta potential, encapsulation efficiency, drug loading rate, drug release in vitro, and stability of PPL NPs were tested. The role of a-HSCs in HCC metastasis was studied by CCK-8, colony formation assay, apoptosis, cellular uptake assay, wound healing assay, and Transwell assay. Western blot was used to detect the related protein expression levels. In vitro and vivo, the acoustic droplet vaporization (ADV) of PPL NPs was tested at different times and LIFU intensities. Biosafety of the PPL NPs was assessed by measuring nude mouse body weight and hematoxylin and eosin (H&E) staining. Results. The results showed that the PPL NPs had good biosafety, with an average particle size of 346.6 ± 62.21 nm and an average zeta potential of -15.23 mV. When the LIFU power is 2.4 W/cm2, it can improve the permeability of cells, further promote the uptake of drugs by cells, and improve the toxicity of drugs. In vitro experiments showed that PPL NPs could inhibit the proliferation of a-HSCs cells, thereby affecting the metastasis of HCC, and were related to the TGFβ-Smad2/3-Snail signaling pathway. Both in vivo and in vitro PPL NPs enhanced ultrasound imaging by LIFU-triggered ADV. Conclusion. The PPL NPs designed and prepared in this study combined with LIFU irradiation could significantly alter the EMT of HCC by inhibiting LX2. Clinically, PPL NPs will also be considered a promising contrast agent due to their ultrasound imaging capabilities.
Collapse
Affiliation(s)
- Xiaomeng Zou
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Tiantian Li
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yingxuan Mao
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Mingwei Zhu
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xi Chen
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jiamei Niu
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Tianxiu Dong
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jian Jiang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xiuhua Yang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
46
|
Chen Y, Wang Z, Wang X, Su M, Xu F, Yang L, Jia L, Zhang Z. Advances in Antitumor Nano-Drug Delivery Systems of 10-Hydroxycamptothecin. Int J Nanomedicine 2022; 17:4227-4259. [PMID: 36134205 PMCID: PMC9482956 DOI: 10.2147/ijn.s377149] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/25/2022] [Indexed: 01/10/2023] Open
Abstract
10-Hydroxycamptothecin (HCPT) is a natural plant alkaloid from Camptotheca that shows potent antitumor activity by targeting intracellular topoisomerase I. However, factors such as instability of the lactone ring and insolubility in water have limited the clinical application of this drug. In recent years, unprecedented advances in biomedical nanotechnology have facilitated the development of nano drug delivery systems. It has been found that nanomedicine can significantly improve the stability and water solubility of HCPT. NanoMedicines with different diagnostic and therapeutic functions have been developed to significantly improve the anticancer effect of HCPT. In this paper, we collected reports on HCPT nanomedicines against tumors in the past decade. Based on current research advances, we dissected the current status and limitations of HCPT nanomedicines development and looked forward to future research directions.
Collapse
Affiliation(s)
- Yukun Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Zhenzhi Wang
- Shaanxi University of Chinese Medicine, Xianyang, 712046, People's Republic of China
| | - Xiaofan Wang
- Department of Oncology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People's Republic of China
| | - Mingliang Su
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Fan Xu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Lian Yang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Zhanxia Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| |
Collapse
|