1
|
Zhang M, Xiang C, Niu R, He X, Luo W, Liu W, Gu R. Liposomes as versatile agents for the management of traumatic and nontraumatic central nervous system disorders: drug stability, targeting efficiency, and safety. Neural Regen Res 2025; 20:1883-1899. [PMID: 39254548 DOI: 10.4103/nrr.nrr-d-24-00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/28/2024] [Indexed: 09/11/2024] Open
Abstract
Various nanoparticle-based drug delivery systems for the treatment of neurological disorders have been widely studied. However, their inability to cross the blood-brain barrier hampers the clinical translation of these therapeutic strategies. Liposomes are nanoparticles composed of lipid bilayers, which can effectively encapsulate drugs and improve drug delivery across the blood-brain barrier and into brain tissue through their targeting and permeability. Therefore, they can potentially treat traumatic and nontraumatic central nervous system diseases. In this review, we outlined the common properties and preparation methods of liposomes, including thin-film hydration, reverse-phase evaporation, solvent injection techniques, detergent removal methods, and microfluidics techniques. Afterwards, we comprehensively discussed the current applications of liposomes in central nervous system diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, traumatic brain injury, spinal cord injury, and brain tumors. Most studies related to liposomes are still in the laboratory stage and have not yet entered clinical trials. Additionally, their application as drug delivery systems in clinical practice faces challenges such as drug stability, targeting efficiency, and safety. Therefore, we proposed development strategies related to liposomes to further promote their development in neurological disease research.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | | | | | | | | | | | | |
Collapse
|
2
|
Aranha MDC, Alencar LMR, Souto EB, Kamei DT, Lopes AM. Dual Chemotherapeutic Loading in Oxalate Transferrin-Conjugated Polymersomes Incorporated into Chitosan Hydrogels for Site-Specific Targeting of Melanoma Cells. Pharmaceuticals (Basel) 2024; 17:1177. [PMID: 39338339 PMCID: PMC11434979 DOI: 10.3390/ph17091177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/24/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
In this work, we developed a smart drug delivery system composed of poly (ethylene glycol)-block-poly (ε-caprolactone) (PEG-PCL)-based polymersomes (Ps) loaded with doxorubicin (DOX) and vemurafenib (VEM). To enhance targeted delivery to malignant melanoma cells, these drug-loaded nanovesicles were conjugated to the oxalate transferrin variant (oxalate Tf) and incorporated into three-dimensional chitosan hydrogels. This innovative approach represents the first application of oxalate Tf for the precision delivery of drug-loaded polymersomes within a semi-solid dosage form based on chitosan hydrogels. These resulting semi-solids exhibited a sustained release profile for both encapsulated drugs. To evaluate their potency, we compared the cytotoxicity of native Tf-Ps with oxalate Tf-Ps. Notably, the oxalate Tf-Ps demonstrated a 3-fold decrease in cell viability against melanoma cells compared to normal cells and were 1.6-fold more potent than native Tf-Ps, indicating the greater potency of this nanoformulation. These findings suggest that dual-drug delivery using an oxalate-Tf-targeting ligand significantly enhances the drug delivery efficiency of Tf-conjugated nanovesicles and offers a promising strategy to overcome the challenge of multidrug resistance in melanoma therapy.
Collapse
Affiliation(s)
- Mariana de C. Aranha
- Department of Biotechnology, Lorena School of Engineering, University of São Paulo (EEL/USP), Lorena 12602-810, Brazil;
| | - Luciana M. R. Alencar
- Laboratory of Biophysics and Nanosystems, Physics Department, Federal University of Maranhão, São Luís 65080-805, Brazil
| | - Eliana B. Souto
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, D04 V1W8 Dublin, Ireland;
| | - Daniel T. Kamei
- Department of Bioengineering, University of California, Los Angeles, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA 90095, USA
| | - André M. Lopes
- Department of Biotechnology, Lorena School of Engineering, University of São Paulo (EEL/USP), Lorena 12602-810, Brazil;
| |
Collapse
|
3
|
Wei Y, Weng X, Wang Y, Yang W. Stimuli-Responsive Polymersomes: Reshaping the Immunosuppressive Tumor Microenvironment. Biomacromolecules 2024; 25:4663-4676. [PMID: 39054960 DOI: 10.1021/acs.biomac.4c00706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
The progression of cancer involves mutations in normal cells, leading to uncontrolled division and tissue destruction, highlighting the complexity of tumor microenvironments (TMEs). Immunotherapy has emerged as a transformative approach, yet the balance between efficacy and safety remains a challenge. Nanoparticles such as polymersomes offer the possibility to precisely target tumors, deliver drugs in a controlled way, effectively modulate the antitumor immunity, and notably reduce side effects. Herein, stimuli-responsive polymersomes, with capabilities for carrying multiple therapeutics, are highlighted for their potential in enhancing antitumor immunity through mechanisms like inducing immunogenic cell death and activating STING (stimulator of interferon genes), etc. The recent progress of utilizing stimuli-responsive polymersomes to reshape the TME is reviewed here. The advantages and limitations to applied stimuli-responsive polymersomes are outlined. Additionally, challenges and future prospects in leveraging polymersomes for cancer therapy are discussed, emphasizing the need for future research and clinical translation.
Collapse
Affiliation(s)
- Yaohua Wei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Xiao Weng
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001 China
| | - Yayun Wang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001 China
| | - Weijing Yang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001 China
| |
Collapse
|
4
|
Marques C, Borchard G, Jordan O. Unveiling the challenges of engineered protein corona from the proteins' perspective. Int J Pharm 2024; 654:123987. [PMID: 38467206 DOI: 10.1016/j.ijpharm.2024.123987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/03/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
It is well known that protein corona affects the "biological identity" of nanoparticles (NPs), which has been seen as both a challenge and an opportunity. Approaches have moved from avoiding protein adsorption to trying to direct it, taking advantage of the formation of a protein corona to favorably modify the pharmacokinetic parameters of NPs. Although promising, the results obtained with engineered NPs still need to be completely understood. While much effort has been put into understanding how the surface of nanomaterials affects protein absorption, less is known about how proteins can affect corona formation due to their specific physicochemical properties. This review addresses this knowledge gap, examining key protein factors influencing corona formation, highlighting current challenges in studying protein-protein interactions, and discussing future perspectives in the field.
Collapse
Affiliation(s)
- Cintia Marques
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel Servet 1211, Geneva, Switzerland; Section of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet 1211, Geneva, Switzerland.
| | - Gerrit Borchard
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel Servet 1211, Geneva, Switzerland; Section of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet 1211, Geneva, Switzerland
| | - Olivier Jordan
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel Servet 1211, Geneva, Switzerland; Section of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet 1211, Geneva, Switzerland
| |
Collapse
|
5
|
Zhang S, Wu X, Liao X, Zhang S. Nanodrug Hijacking Blood Transferrin for Ferroptosis-Mediated Cancer Treatment. J Am Chem Soc 2024; 146:8567-8575. [PMID: 38489761 DOI: 10.1021/jacs.4c00395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Ferroptosis as a promising method of cancer treatment heavily relies on the intracellular iron ion level. Herein, a new iron-supplement nanodrug was developed by conjugating transferrin-homing peptide T10 on the surface of cross-linked lipoic acid vesicles (T10@cLAV), which could hijack blood transferrin (Tf) and specifically deliver it to tumor cells to elevate the Fe2+ level. Meanwhile, the intracellular degradation product of cLAV, dihydrolipoic acid, could regenerate Fe2+ to further boost the ferroptosis. The results disclosed that T10@cLAV achieved tumor inhibition comparable to that of cisplatin at a dose as low as 5 mg/kg in the HeLa tumor-bearing nude mice model and caused no toxicity at the dose up to 300 mg/kg. This tactful iron-supplement strategy of hijacking blood Tf is superior to the current strategies: one is the induction of intracellular ferritin degradation, which is limited by the low content of ferritin, and the other is the delivery of iron-based materials, which easily causes adverse effects.
Collapse
Affiliation(s)
- Shuyue Zhang
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 610064 Chengdu, China
| | - Xiao Wu
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 610064 Chengdu, China
| | - Xiaoming Liao
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 610064 Chengdu, China
| | - Shiyong Zhang
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 610064 Chengdu, China
| |
Collapse
|
6
|
Skowicki M, Hürlimann D, Tarvirdipour S, Kyropoulou M, Schoenenberger CA, Gerber-Lemaire S, Palivan CG. FAP Targeting of Photosensitizer-Loaded Polymersomes for Increased Light-Activated Cell Killing. Biomacromolecules 2024; 25:754-766. [PMID: 38267014 PMCID: PMC10865352 DOI: 10.1021/acs.biomac.3c00943] [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: 09/06/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 01/26/2024]
Abstract
As current chemo- and photodynamic cancer therapies are associated with severe side effects due to a lack of specificity and to systemic toxicity, innovative solutions in terms of targeting and controlled functionality are in high demand. Here, we present the development of a polymersome nanocarrier equipped with targeting molecules and loaded with photosensitizers for efficient uptake and light-activated cell killing. Polymersomes were self-assembled in the presence of photosensitizers from a mixture of nonfunctionalized and functionalized PDMS-b-PMOXA diblock copolymers, the latter designed for coupling with targeting ligands. By encapsulation inside the polymersomes, the photosensitizer Rose Bengal was protected, and its uptake into cells was mediated by the nanocarrier. Inhibitor of fibroblast activation protein α (FAPi), a ligand for FAP, was attached to the polymersomes' surface and improved their uptake in MCF-7 breast cancer cells expressing relatively high levels of FAP on their surface. Once internalized by MCF-7, irradiation of Rose Bengal-loaded FAPi-polymersomes generated reactive oxygen species at levels high enough to induce cell death. By combining photosensitizer encapsulation and specific targeting, polymersomes represent ideal candidates as therapeutic nanocarriers in cancer treatment.
Collapse
Affiliation(s)
- Michal Skowicki
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
- NCCR-Molecular
Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Dimitri Hürlimann
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
- NCCR-Molecular
Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Shabnam Tarvirdipour
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
| | - Myrto Kyropoulou
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
- NCCR-Molecular
Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cora-Ann Schoenenberger
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
- NCCR-Molecular
Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Sandrine Gerber-Lemaire
- Group
for Functionalized Biomaterials, Institute of Chemical Sciences and
Engineering, Ecole Polytechnique Fédérale
de Lausanne, CH-1015 Lausanne, Switzerland
| | - Cornelia G. Palivan
- Department
of Chemistry, University of Basel, BPR 1096, Mattenstrasse 22, 4058 Basel, Switzerland
- NCCR-Molecular
Systems Engineering, BPR 1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| |
Collapse
|
7
|
Guo F, Du Y, Wang Y, Wang M, Wang L, Yu N, Luo S, Wu F, Yang G. Targeted drug delivery systems for matrix metalloproteinase-responsive anoparticles in tumor cells: A review. Int J Biol Macromol 2024; 257:128658. [PMID: 38065446 DOI: 10.1016/j.ijbiomac.2023.128658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/26/2024]
Abstract
Nanodrug delivery systems based on tumor microenvironment responses have shown excellent performance in tumor-targeted therapy, given their unique targeting and drug-release characteristics. Matrix metalloproteinases (MMPs) have been widely explored owing to their high specificity and expression in various tumor microenvironments. The design of an enzyme-sensitive nanodelivery system using MMPs as targeted receptors could markedly improve the performance of drug targeting. The current review focuses on the development and application of MMP-responsive drug carriers, and summarizes the classification of single- and multi-target nanocarriers based on their MMP responsiveness. The potential applications and challenges of this nanodrug delivery system are discussed to provide a reference for designing high-performance nanodrug delivery systems.
Collapse
Affiliation(s)
- Fangyuan Guo
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yinzhou Du
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yujia Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Mengqi Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Lianyi Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Nan Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shuai Luo
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Fang Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Gensheng Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
8
|
Abdellatif AAH, Alshubrumi AS, Younis MA. Targeted Nanoparticles: the Smart Way for the Treatment of Colorectal Cancer. AAPS PharmSciTech 2024; 25:23. [PMID: 38267656 DOI: 10.1208/s12249-024-02734-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024] Open
Abstract
Colorectal cancer (CRC) is a widespread cancer that starts in the digestive tract. It is the third most common cause of cancer deaths around the world. The World Health Organization (WHO) estimates an expected death toll of over 1 million cases annually. The limited therapeutic options as well as the drawbacks of the existing therapies necessitate the development of non-classic treatment approaches. Nanotechnology has led the evolution of valuable drug delivery systems thanks to their ability to control drug release and precisely target a wide variety of cancers. This has also been extended to the treatment of CRC. Herein, we shed light on the pertinent research that has been performed on the potential applications of nanoparticles in the treatment of CRC. The various types of nanoparticles in addition to their properties, applications, targeting approaches, merits, and demerits are discussed. Furthermore, innovative therapies for CRC, including gene therapies and immunotherapies, are also highlighted. Eventually, the research gaps, the clinical potential of such delivery systems, and a future outlook on their development are inspired.
Collapse
Affiliation(s)
- Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, 51452, Buraydah, Al Qassim, Saudi Arabia.
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt.
| | | | - Mahmoud A Younis
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt.
| |
Collapse
|
9
|
Song Q, Zheng Y, Zhong G, Wang S, He C, Li M. Application of Nanoparticles in the Diagnosis and Treatment of Colorectal Cancer. Anticancer Agents Med Chem 2024; 24:1305-1326. [PMID: 39129164 PMCID: PMC11497148 DOI: 10.2174/0118715206323900240807110122] [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: 05/15/2024] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024]
Abstract
Colorectal cancer is a common malignant tumor with high morbidity and mortality rates, imposing a huge burden on both patients and the healthcare system. Traditional treatments such as surgery, chemotherapy and radiotherapy have limitations, so finding more effective diagnostic and therapeutic tools is critical to improving the survival and quality of life of colorectal cancer patients. While current tumor targeting research mainly focuses on exploring the function and mechanism of molecular targets and screening for excellent drug targets, it is crucial to test the efficacy and mechanism of tumor cell therapy that targets these molecular targets. Selecting the appropriate drug carrier is a key step in effectively targeting tumor cells. In recent years, nanoparticles have gained significant interest as gene carriers in the field of colorectal cancer diagnosis and treatment due to their low toxicity and high protective properties. Nanoparticles, synthesized from natural or polymeric materials, are NM-sized particles that offer advantages such as low toxicity, slow release, and protection of target genes during delivery. By modifying nanoparticles, they can be targeted towards specific cells for efficient and safe targeting of tumor cells. Numerous studies have demonstrated the safety, efficiency, and specificity of nanoparticles in targeting tumor cells, making them a promising gene carrier for experimental and clinical studies. This paper aims to review the current application of nanoparticles in colorectal cancer diagnosis and treatment to provide insights for targeted therapy for colorectal cancer while also highlighting future prospects for nanoparticle development.
Collapse
Affiliation(s)
- Qiuyu Song
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yifeng Zheng
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guoqiang Zhong
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shanping Wang
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chengcheng He
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingsong Li
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
Chen Q, Han X, Liu L, Duan Y, Chen Y, Shi L, Lin Q, Shen L. Multifunctional Polymer Vesicles for Synergistic Antibiotic-Antioxidant Treatment of Bacterial Keratitis. Biomacromolecules 2023; 24:5230-5244. [PMID: 37733485 DOI: 10.1021/acs.biomac.3c00754] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
As an acute ophthalmic infection, bacterial keratitis (BK) can lead to severe visual morbidity, such as corneal perforation, intraocular infection, and permanent corneal opacity, if rapid and effective treatments are not available. In addition to eradicating pathogenic bacteria, protecting corneal tissue from oxidative damage and promoting wound healing by relieving inflammation are equally critical for the efficient treatment of BK. Besides, it is very necessary to improve the bioavailability of drugs by enhancing the ocular surface adhesion and corneal permeability. In this investigation, therefore, a synergistic antibiotic-antioxidant treatment of BK was achieved based on multifunctional block copolymer vesicles, within which ciprofloxacin (CIP) was simultaneously encapsulated during the self-assembly. Due to the phenylboronic acid residues in the corona layer, these vesicles exhibited enhanced muco-adhesion, deep corneal epithelial penetration, and bacteria-targeting, which facilitated the drug delivery to corneal bacterial infection sites. Additionally, the abundant thioether moieties in the hydrophobic membrane enabled the vesicles to both have ROS-scavenging capacity and accelerated CIP release at the inflammatory corneal tissue. In vivo experiments on a mice model demonstrated that the multifunctional polymer vesicles achieved efficient treatment of BK, owing to the enhanced corneal adhesion and penetration, bacteria targeting, ROS-triggered CIP release, and the combined antioxidant-antibiotic therapy. This synergistic strategy holds great potential in the treatment of BK and other diseases associated with bacterial infections.
Collapse
Affiliation(s)
- Qiumeng Chen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaopeng Han
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| | - Lu Liu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| | - Yong Duan
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| | - Yifei Chen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Quankui Lin
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| | - Liangliang Shen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
11
|
Kayani A, Raza A, Si J, Dutta D, Zhou Q, Ge Z. Polymersome Membrane Engineering with Active Targeting or Controlled Permeability for Responsive Drug Delivery. Biomacromolecules 2023; 24:4622-4645. [PMID: 37870458 DOI: 10.1021/acs.biomac.3c00839] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Polymersomes have been extensively investigated for drug delivery as nanocarriers for two decades due to a series of advantages including high stability under physiological conditions, simultaneous encapsulation of hydrophilic and hydrophobic drugs inside inner cavities and membranes, respectively, and facile adjustment of membrane and surface properties, as well as controlled drug release through incorporation of stimuli-responsive components. Despite these features, polymersome nanocarriers frequently suffer from nontargeting delivery and poor membrane permeability. In recent years, polymersomes have been functionalized for more efficient drug delivery. The surface shells were explored to be modified with diverse active targeting groups to improve disease-targeting delivery. The membrane permeability of the polymersomes was adjusted by incorporation of the stimuli-responsive components for smart controlled transportation of the encapsulated drugs. Therefore, being the polymersome-biointerface, tailorable properties can be introduced by its carefully modulated engineering. This review elaborates on the role of polymersome membranes as a platform to incorporate versatile features. First, we discuss how surface functionalization facilitates the directional journey to the targeting sites toward specific diseases, cells, or intracellular organelles via active targeting. Moreover, recent advances in the past decade related to membrane permeability to control drug release are also summarized. We finally discuss future development to promote polymersomes as in vivo drug delivery nanocarriers.
Collapse
Affiliation(s)
- Anum Kayani
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Arsalan Raza
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Jiale Si
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Debabrata Dutta
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Qinghao Zhou
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Zhishen Ge
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| |
Collapse
|
12
|
Choudhury H, Pandey M, Saravanan V, Mun ATY, Bhattamisra SK, Parikh A, Garg S, Gorain B. Recent progress of targeted nanocarriers in diagnostic, therapeutic, and theranostic applications in colorectal cancer. BIOMATERIALS ADVANCES 2023; 153:213556. [PMID: 37478770 DOI: 10.1016/j.bioadv.2023.213556] [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: 04/18/2023] [Revised: 06/28/2023] [Accepted: 07/06/2023] [Indexed: 07/23/2023]
Abstract
Cancer at the lower end of the digestive tract, colorectal cancer (CRC), starts with asymptomatic polyps, which can be diagnosed as cancer at a later stage. It is the fourth leading cause of malignancy-associated mortality worldwide. Despite progress in conventional treatment strategies, the possibility to overcome the mortality and morbidity issues with the enhancement of the lifespan of CRC patients is limited. With the advent of nanocarrier-based drug delivery systems, a promising revolution has been made in diagnosis, treatment, and theranostic purposes for cancer management. Herein, we reviewed the progress of miniaturized nanocarriers, such as liposomes, niosomes, solid lipid nanoparticles, micelles, and polymeric nanoparticles, employed in passive and active targeting and their role in theranostic applications in CRC. With this novel scope, the diagnosis and treatment of CRC have proceeded to the forefront of innovation, where specific characteristics of the nanocarriers, such as processability, flexibility in developing precise architecture, improved circulation, site-specific delivery, and rapid response, facilitate the management of cancer patients. Furthermore, surface-engineered technologies for the nanocarriers could involve receptor-mediated deliveries towards the overexpressed receptors on the CRC microenvironment. Moreover, the potential of clinical translation of these targeted miniaturized formulations as well as the possible limitations and barriers that could impact this translation into clinical practice were highlighted. The advancement of these newest developments in clinical research and progress into the commercialization stage gives hope for a better tomorrow.
Collapse
Affiliation(s)
- Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Manisha Pandey
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia.
| | - Vilashini Saravanan
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Amanda Tan Yee Mun
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Subrat Kumar Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Ankit Parikh
- Centre for Pharmaceutical Innovation (CPI), Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Sanjay Garg
- Centre for Pharmaceutical Innovation (CPI), Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India.
| |
Collapse
|
13
|
Kansız S, Elçin YM. Advanced liposome and polymersome-based drug delivery systems: Considerations for physicochemical properties, targeting strategies and stimuli-sensitive approaches. Adv Colloid Interface Sci 2023; 317:102930. [PMID: 37290380 DOI: 10.1016/j.cis.2023.102930] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 06/10/2023]
Abstract
Liposomes and polymersomes are colloidal vesicles that are self-assembled from lipids and amphiphilic polymers, respectively. Because of their ability to encapsulate both hydrophilic and hydrophobic therapeutics, they are of great interest in drug delivery research. Today, the applications of liposomes and polymersomes have expanded to a wide variety of complex therapeutic molecules, including nucleic acids, proteins and enzymes. Thanks to their chemical versatility, they can be tailored to different drug delivery applications to achieve maximum therapeutic index. This review article evaluates liposomes and polymersomes from a perspective that takes into account the physical and biological barriers that reduce the efficiency of the drug delivery process. In this context, the design approaches of liposomes and polymersomes are discussed with representative examples in terms of their physicochemical properties (size, shape, charge, mechanical), targeting strategies (passive and active) and response to different stimuli (pH, redox, enzyme, temperature, light, magnetic field, ultrasound). Finally, the challenges limiting the transition from laboratory to practice, recent clinical developments, and future perspectives are addressed.
Collapse
Affiliation(s)
- Seyithan Kansız
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Department of Chemistry, Ankara, Turkey
| | - Yaşar Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Department of Chemistry, Ankara, Turkey; Biovalda Health Technologies, Inc., Ankara, Turkey.
| |
Collapse
|
14
|
Recent applications of phase-change materials in tumor therapy and theranostics. BIOMATERIALS ADVANCES 2023; 147:213309. [PMID: 36739784 DOI: 10.1016/j.bioadv.2023.213309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Phase-change materials (PCMs) are a type of special material which can store and release a large amount of thermal energy without any significant temperature change. They are emerging in recent years as a promising functional material in tumor therapy and theranostics due to their accurate responses to the temperature variations, biocompatibility and low toxicity. In this review, we will introduce the main types of PCMs and their desirable physiochemical properties for biomedical applications, and highlight the recent progress of PCM's applications in the modulated release of antitumor drugs, with special attentions paid to various ways to initiate temperature-dependent phase change, the concomitant thermal therapy and its combination with or activation of other therapies, particularly unconventional therapies. We will also summarize PCM's recent applications in tumor theranostics, where both drugs and imaging probes are delivered by PCMs for controlled drug release and imaging-guided therapy. Finally, the future perspectives and potential limitations of harnessing PCMs in tumor therapy will be discussed.
Collapse
|
15
|
Duan S, Sun F, Qiao P, Zhu Z, Geng M, Gong X, Li Y, Yao H. Detachable Dual-Targeting Nanoparticles for Improving the Antitumor Effect by Extracellular Matrix Depletion. ACS Biomater Sci Eng 2023; 9:1437-1449. [PMID: 36795746 DOI: 10.1021/acsbiomaterials.2c01179] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
In the tumor microenvironment (TME), the extracellular matrix (ECM) produced by cancer-associated fibroblasts (CAFs) forms a dense barrier that prevents nanodrugs from penetrating into deep tumor sites, leading to unsatisfactory therapeutic effects. Recently, it has been found that ECM depletion and using small-sized nanoparticles are effective strategies. Herein, we reported a detachable dual-targeting nanoparticle (HA-DOX@GNPs-Met@HFn) based on reducing ECM for enhancing penetration. When these nanoparticles reached the tumor site, the nanoparticles were divided into two parts in response to matrix metalloproteinase-2 overexpressed in TME, causing a decrease in the nanoparticle size from about 124 to 36 nm. One part was Met@HFn, which was detached from the surface of gelatin nanoparticles (GNPs), which effectively targeted tumor cells and released metformin (Met) under acidic conditions. Then, Met downregulated the expression of the transforming growth factor β by the adenosine monophosphate-activated protein kinase pathway to inhibit the activity of CAFs, thereby suppressing the production of ECM including α-smooth muscle actin and collagen I. The other was the small-sized hyaluronic acid-modified doxorubicin prodrug with autonomous targeting ability, which was gradually released from GNPs and internalized into deeper tumor cells. Intracellular hyaluronidases triggered the release of doxorubicin (DOX), which killed tumor cells by inhibiting DNA synthesis. The combination of size transformation and ECM depletion enhanced the penetration and accumulation of DOX in solid tumors. Therefore, the tumor chemotherapy effect was greatly improved.
Collapse
Affiliation(s)
- Songchao Duan
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Fangfang Sun
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Pan Qiao
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Zhihui Zhu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Meilin Geng
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Xiaobao Gong
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Ying Li
- Department of Pharmacy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Hanchun Yao
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, 100 Science Road, Zhengzhou 450001, China
| |
Collapse
|
16
|
Hung J, Perez SM, Dasa SSK, Hall SP, Heckert DB, Murphy BP, Crawford HC, Kelly KA, Brinton LT. A Bitter Taste Receptor as a Novel Molecular Target on Cancer-Associated Fibroblasts in Pancreatic Ductal Adenocarcinoma. Pharmaceuticals (Basel) 2023; 16:389. [PMID: 36986488 PMCID: PMC10058050 DOI: 10.3390/ph16030389] [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: 12/03/2022] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) execute diverse and complex functions in cancer progression. While reprogramming the crosstalk between CAFs and cancer epithelial cells is a promising avenue to evade the adverse effects of stromal depletion, drugs are limited by their suboptimal pharmacokinetics and off-target effects. Thus, there is a need to elucidate CAF-selective cell surface markers that can improve drug delivery and efficacy. Here, functional proteomic pulldown with mass spectrometry was used to identify taste receptor type 2 member 9 (TAS2R9) as a CAF target. TAS2R9 target characterization included binding assays, immunofluorescence, flow cytometry, and database mining. Liposomes conjugated to a TAS2R9-specific peptide were generated, characterized, and compared to naked liposomes in a murine pancreatic xenograft model. Proof-of-concept drug delivery experiments demonstrate that TAS2R9-targeted liposomes bind with high specificity to TAS2R9 recombinant protein and exhibit stromal colocalization in a pancreatic cancer xenograft model. Furthermore, the delivery of a CXCR2 inhibitor by TAS2R9-targeted liposomes significantly reduced cancer cell proliferation and constrained tumor growth through the inhibition of the CXCL-CXCR2 axis. Taken together, TAS2R9 is a novel cell-surface CAF-selective target that can be leveraged to facilitate small-molecule drug delivery to CAFs, paving the way for new stromal therapies.
Collapse
Affiliation(s)
- Jessica Hung
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | | - Siva Sai Krishna Dasa
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | - Howard C. Crawford
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, MI 48202, USA
| | - Kimberly A. Kelly
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- ZielBio Inc., Charlottesville, VA 22902, USA
| | - Lindsey T. Brinton
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- ZielBio Inc., Charlottesville, VA 22902, USA
| |
Collapse
|
17
|
Zhang Q, Liu N, Wang J, Liu Y, Wang K, Zhang J, Pan X. The Recent Advance of Cell-Penetrating and Tumor-Targeting Peptides as Drug Delivery Systems Based on Tumor Microenvironment. Mol Pharm 2023; 20:789-809. [PMID: 36598861 DOI: 10.1021/acs.molpharmaceut.2c00629] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cancer has become the primary reason for industrial countries death. Although first-line treatments have achieved remarkable results in inhibiting tumors, they could have serious side effects because of insufficient selectivity. Therefore, specific localization of tumor cells is currently the main desire for cancer treatment. In recent years, cell-penetrating peptides (CPPs), as a kind of promising delivery vehicle, have attracted much attention because they mediate the high-efficiency import of large quantities of cargos in vivo and vitro. Unfortunately, the poor targeting of CPPs is still a barrier to their clinical application. In order to solve this problem, researchers use the various characteristics of tumor microenvironment and multiple receptors to improve the specificity toward tumors. This review focuses on the characteristics of the tumor microenvironment, and introduces the development of strategies and peptides based on these characteristics as drug delivery system in the tumor-targeted therapy.
Collapse
Affiliation(s)
- Qingqing Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Nanxin Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuying Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Kai Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
18
|
Alimardani V, Rahiminezhad Z, DehghanKhold M, Farahavar G, Jafari M, Abedi M, Moradi L, Niroumand U, Ashfaq M, Abolmaali SS, Yousefi G. Nanotechnology-based cell-mediated delivery systems for cancer therapy and diagnosis. Drug Deliv Transl Res 2023; 13:189-221. [PMID: 36074253 DOI: 10.1007/s13346-022-01211-9] [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] [Accepted: 07/11/2022] [Indexed: 12/13/2022]
Abstract
The global prevalence of cancer is increasing, necessitating new additions to traditional treatments and diagnoses to address shortcomings such as ineffectiveness, complications, and high cost. In this context, nano and microparticulate carriers stand out due to their unique properties such as controlled release, higher bioavailability, and lower toxicity. Despite their popularity, they face several challenges including rapid liver uptake, low chemical stability in blood circulation, immunogenicity concerns, and acute adverse effects. Cell-mediated delivery systems are important topics to research because of their biocompatibility, biodegradability, prolonged delivery, high loading capacity, and targeted drug delivery capabilities. To date, a variety of cells including blood, immune, cancer, and stem cells, sperm, and bacteria have been combined with nanoparticles to develop efficient targeted cancer delivery or diagnosis systems. The review paper aimed to provide an overview of the potential applications of cell-based delivery systems in cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Vahid Alimardani
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Rahiminezhad
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahvash DehghanKhold
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ghazal Farahavar
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Jafari
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Abedi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Moradi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Uranous Niroumand
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ashfaq
- University Centre for Research & Development (UCRD), Chandigarh University, Gharaun, Mohali, 140413, Punjab, India. .,Department of Biotechnology, Chandigarh University, Gharaun, Mohali, 140413, Punjab, India.
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran. .,Center for Drug Delivery in Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Gholamhossein Yousefi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran. .,Center for Drug Delivery in Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
19
|
Huang L, Li W, Lu Y, Ju Q, Ouyang M. Iron metabolism in colorectal cancer. Front Oncol 2023; 13:1098501. [PMID: 36910614 PMCID: PMC9992732 DOI: 10.3389/fonc.2023.1098501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Iron, as one of the essential trace elements in the human body, is involved in a wide range of critical biochemical reactions and physiological processes, including the maintenance of the normal cell cycle, mitochondrial function, nucleotide metabolism, and immune response. In this context, iron is naturally associated with cancer occurrence. Cellular iron deficiency can induce apoptosis, however, iron can also engage in potentially harmful reactions that produce free radicals because of its capacity to gain and lose electrons. Studies suggest that dietary iron, particularly heme iron, may be one of the leading causes of colorectal cancer (CRC). Moreover, patients with CRC have abnormal iron absorption, storage, utilization, and exportation. Therefore, iron is crucial for the development and progression of CRC. Elaborating on the alterations in iron metabolism during the onset and advancement of CRC would help to further explain the role and mechanism of iron inside the body. Thus, we reviewed the alterations in numerous iron metabolism-related molecules and their roles in CRC, which may provide new clues between iron metabolism and CRC.
Collapse
Affiliation(s)
- Luji Huang
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Wangji Li
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan Lu
- Good Clinical Practice (GCP) Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Guangdong, China
| | - Qinuo Ju
- Guangdong Country Garden School, Shunde, Foshan, Guangdong, China
| | - Manzhao Ouyang
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| |
Collapse
|
20
|
Yadav DN, Ali MS, Thanekar AM, Pogu SV, Rengan AK. Recent Advancements in the Design of Nanodelivery Systems of siRNA for Cancer Therapy. Mol Pharm 2022; 19:4506-4526. [PMID: 36409653 DOI: 10.1021/acs.molpharmaceut.2c00811] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
RNA interference (RNAi) has increased the possibility of restoring RNA drug targets for cancer treatment. Small interfering RNA (siRNA) is a promising therapeutic RNAi tool that targets the defective gene by inhibiting its mRNA expression and stopping its translation. However, siRNAs have flaws like poor intracellular trafficking, RNase degradation, rapid kidney filtration, off-targeting, and toxicity, which limit their therapeutic efficiency. Nanocarriers (NCs) have been designed to overcome such flaws and increase antitumor activity. Combining siRNA and anticancer drugs can give synergistic effects in cancer cells, making them a significant gene-modification tool in cancer therapy. Our discussion of NCs-mediated siRNA delivery in this review includes their mechanism, limitations, and advantages in comparison with naked siRNA delivery. We will also discuss organic NCs (polymers and lipids) and inorganic NCs (quantum dots, carbon nanotubes, and gold) that have been reported for extensive delivery of therapeutic siRNA to tumor sites. Finally, we will conclude by discussing the studies based on organic and inorganic NCs-mediated siRNA drug delivery systems conducted in the years 2020 and 2021.
Collapse
Affiliation(s)
- Dokkari Nagalaxmi Yadav
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| | - Mohammad Sadik Ali
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| | | | - Sunil Venkanna Pogu
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| |
Collapse
|
21
|
Strategies to improve drug penetration into tumor microenvironment by nanoparticles: focus on nanozymes. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
22
|
IL-11Rα-targeted nanostrategy empowers chemotherapy of relapsed and patient-derived osteosarcoma. J Control Release 2022; 350:460-470. [PMID: 36041590 DOI: 10.1016/j.jconrel.2022.08.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/22/2022]
Abstract
Osteosarcoma (OS) is a rare but frequently lethal bone malignancy in children and adolescents. The adjuvant chemotherapy with doxorubicin (Dox) and cisplatin remains a mainstream clinical practice though it affords only limited clinical benefits due to low tumor deposition, dose-limiting toxicity and high rate of relapse/metastasis. Here, taking advantage of high IL-11Rα expression in the OS patients, we installed IL-11Rα specific peptide (sequence: CGRRAGGSC) onto redox-responsive polymersomes encapsulating Dox (IL11-PDox) to boost the specificity and anti-OS efficacy of chemotherapy. Of note, IL-11Rα peptide at a density of 20% greatly augmented the internalization, apoptotic activity, and migration inhibition of Dox in IL-11Rα-overexpressing 143B OS cells. The active targeting effect of IL-11-PDox was supported in orthotopic and relapsed 143B OS models, as shown by striking repression of tumor growth and lung metastasis and substantial survival benefits over free Dox control. We further verified that IL11-PDox could effectively inhibit patient-derived OS xenografts. IL-11Rα-targeted nanodelivery of chemotherapeutics provides a potential therapeutic strategy for advanced osteosarcoma.
Collapse
|
23
|
Liu J, Zhang L, Zhao D, Yue S, Sun H, Ni C, Zhong Z. Polymersome-stabilized doxorubicin-lipiodol emulsions for high-efficacy chemoembolization therapy. J Control Release 2022; 350:122-131. [PMID: 35973474 DOI: 10.1016/j.jconrel.2022.08.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/03/2022] [Accepted: 08/10/2022] [Indexed: 02/08/2023]
Abstract
Transarterial chemoembolization (TACE) with free doxorubicin-lipiodol emulsions (free DOX/L) is a favored clinical treatment for advanced hepatocellular carcinoma (HCC) patients ineligible for radical therapies; however, its inferior colloidal stability not only greatly reduces its tumor retention but also hastens drug release into blood circulation, leading to suboptimal clinical outcomes. Here, we find that disulfide-crosslinked polymersomes carrying doxorubicin (Ps-DOX) form super-stable and homogenous water-in-oil microemulsions with lipiodol (Ps-DOX/L). Ps-DOX/L microemulsions had tunable sizes ranging from 14 to 44 μm depending on the amount of Ps-DOX, were stable over 2 months storage as well as centrifugation, and exhibited nearly zero-order DOX release within 15 days. Of note, Ps-DOX induced 2.3-13.4 fold better inhibitory activity in all tested rat, murine and human liver tumor cells than free DOX likely due to its efficient redox-triggered intracellular drug release. Interestingly, transarterial administration of Ps-DOX/L microemulsions in orthotopic rat N1S1 syngeneic HCC model showed minimal systemic DOX exposure, high and long hepatic DOX retention, complete tumor elimination, effective inhibition of angiogenesis, and depleted adverse effects, significantly outperforming clinically used free DOX/L emulsions. This smart polymersome stabilization of doxorubicin-lipiodol microemulsions provides a novel TACE strategy for advanced tumors.
Collapse
Affiliation(s)
- Jingyi Liu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Lei Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215123, PR China
| | - Dongxu Zhao
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215123, PR China
| | - Shujing Yue
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Huanli Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| | - Caifang Ni
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215123, PR China.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
24
|
Guo Y, Wang M, Zou Y, Jin L, Zhao Z, Liu Q, Wang S, Li J. Mechanisms of chemotherapeutic resistance and the application of targeted nanoparticles for enhanced chemotherapy in colorectal cancer. J Nanobiotechnology 2022; 20:371. [PMID: 35953863 PMCID: PMC9367166 DOI: 10.1186/s12951-022-01586-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/04/2022] [Indexed: 11/10/2022] Open
Abstract
Colorectal cancer is considered one of the major malignancies that threaten the lives and health of people around the world. Patients with CRC are prone to post-operative local recurrence or metastasis, and some patients are advanced at the time of diagnosis and have no chance for complete surgical resection. These factors make chemotherapy an indispensable and important tool in treating CRC. However, the complex composition of the tumor microenvironment and the interaction of cellular and interstitial components constitute a tumor tissue with high cell density, dense extracellular matrix, and high osmotic pressure, inevitably preventing chemotherapeutic drugs from entering and acting on tumor cells. As a result, a novel drug carrier system with targeted nanoparticles has been applied to tumor therapy. It can change the physicochemical properties of drugs, facilitate the crossing of drug molecules through physiological and pathological tissue barriers, and increase the local concentration of nanomedicines at lesion sites. In addition to improving drug efficacy, targeted nanoparticles also reduce side effects, enabling safer and more effective disease diagnosis and treatment and improving bioavailability. In this review, we discuss the mechanisms by which infiltrating cells and other stromal components of the tumor microenvironment comprise barriers to chemotherapy in colorectal cancer. The research and application of targeted nanoparticles in CRC treatment are also classified.
Collapse
Affiliation(s)
- Yu Guo
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Min Wang
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Yongbo Zou
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Longhai Jin
- Department of Radiology, Jilin University Second Hospital, Changchun, 130000, China
| | - Zeyun Zhao
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Qi Liu
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Shuang Wang
- Department of the Dermatology, Jilin University Second Hospital, Changchun, 130000, China.
| | - Jiannan Li
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China.
| |
Collapse
|
25
|
Song D, Xu Q. Engineering a Nano/Biointerface for Cell and Organ-Selective Drug Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:9092-9098. [PMID: 35852946 DOI: 10.1021/acs.langmuir.2c01609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The field of nanomedicine has rapidly grown in the past decades. Although a few nanomedicines are available in the market for clinical use, it is still challenging to develop nanomedicine targeting tissues beyond the liver. It has been recognized that even though the nanoparticles are modified with targeting ligands, the formation of a protein corona on the surface of nanoparticles in the biological fluids results in limited progress in nanoparticle-based drug delivery to specific cells or tissues. In this Perspective, we will discuss the role of surface properties in determining the formation of the protein corona and summarize the recent progress in engineering the nano/bio interface for protein-corona-mediated cell- and organ-selective drug delivery. Moreover, current challenges in the field and insights into designing new strategies for targeting drug delivery with a better understanding of the protein corona will be discussed.
Collapse
Affiliation(s)
- Donghui Song
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
26
|
Haider M, Zaki KZ, El Hamshary MR, Hussain Z, Orive G, Ibrahim HO. Polymeric nanocarriers: A promising tool for early diagnosis and efficient treatment of colorectal cancer. J Adv Res 2022; 39:237-255. [PMID: 35777911 PMCID: PMC9263757 DOI: 10.1016/j.jare.2021.11.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/03/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most prevalent type of cancer for incidence and second for mortality worldwide. Late diagnosis and inconvenient and expensive current diagnostic tools largely contribute to the progress of the disease. The use of chemotherapy in the management of CRC significantly reduces tumor growth, metastasis, and morbidity rates. However, poor solubility, low cellular uptake, nonspecific distribution, multiple drug resistance and unwanted adverse effects are still among the major drawbacks of chemotherapy that limit its clinical significance in the treatment of CRC. Owing to their remarkable advantages over conventional therapies, the use of nanotechnology-based delivery systems especially polymeric nanocarriers (PNCs) has revolutionized many fields including disease diagnosis and drug delivery. AIM OF REVIEW In this review, we shed the light on the current status of using PNCs in the diagnosis and treatment of CRC with a special focus on targeting strategies, surface modifications and safety concerns for different types of PNCs in colonic cancer delivery. KEY SCIENTIFIC CONCEPTS OF REVIEW The review explores the current progress on the use of PNCs in the diagnosis and treatment of CRC with a special focus on the role of PNCs in improvement of cellular uptake, drug targeting and co-delivery of chemotherapeutic agents. Possible toxicity and biocompatibility issues related to the use of PNCs and imitations and future recommendation for the use of those smart carriers in the diagnosis and treatment of CRC are also discussed.
Collapse
Affiliation(s)
- Mohamed Haider
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 71526, Egypt.
| | - Khaled Zaki Zaki
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mariam Rafat El Hamshary
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Zahid Hussain
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Haidy Osama Ibrahim
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
27
|
Hou S, Hasnat M, Chen Z, Liu Y, Faran Ashraf Baig MM, Liu F, Chen Z. Application Perspectives of Nanomedicine in Cancer Treatment. Front Pharmacol 2022; 13:909526. [PMID: 35860027 PMCID: PMC9291274 DOI: 10.3389/fphar.2022.909526] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer is a disease that seriously threatens human health. Based on the improvement of traditional treatment methods and the development of new treatment modes, the pattern of cancer treatment is constantly being optimized. Nanomedicine plays an important role in these evolving tumor treatment modalities. In this article, we outline the applications of nanomedicine in three important tumor-related fields: chemotherapy, gene therapy, and immunotherapy. According to the current common problems, such as poor targeting of first-line chemotherapy drugs, easy destruction of nucleic acid drugs, and common immune-related adverse events in immunotherapy, we discuss how nanomedicine can be combined with these treatment modalities, provide typical examples, and summarize the advantages brought by the application of nanomedicine.
Collapse
Affiliation(s)
- Shanshan Hou
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
| | - Muhammad Hasnat
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Ziwei Chen
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
| | - Yinong Liu
- Hospital Laboratory of Nangjing Lishui People’s Hospital, Nangjing, China
| | - Mirza Muhammad Faran Ashraf Baig
- Laboratory of Biomedical Engineering for Novel Bio-functional, and Pharmaceutical Nanomaterials, Prince Philip Dental Hospital, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Fuhe Liu
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
- *Correspondence: Zelong Chen, ; Fuhe Liu,
| | - Zelong Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Province Engineering Research Center of Artificial Intelligence and Internet of Things Wise Medical, Zhengzhou, China
- *Correspondence: Zelong Chen, ; Fuhe Liu,
| |
Collapse
|
28
|
Xiao Q, Zoulikha M, Qiu M, Teng C, Lin C, Li X, Sallam MA, Xu Q, He W. The effects of protein corona on in vivo fate of nanocarriers. Adv Drug Deliv Rev 2022; 186:114356. [PMID: 35595022 DOI: 10.1016/j.addr.2022.114356] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/01/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022]
Abstract
With the emerging advances in utilizing nanocarriers for biomedical applications, a molecular-level understanding of the in vivo fate of nanocarriers is necessary. After administration into human fluids, nanocarriers can attract proteins onto their surfaces, forming an assembled adsorption layer called protein corona (PC). The formed PC can influence the physicochemical properties and subsequently determine nanocarriers' biological behaviors. Therefore, an in-depth understanding of the features and effects of the PC on the nanocarriers' surface is the first and most important step towards controlling their in vivo fate. This review introduces fundamental knowledge such as the definition, formation, composition, conformation, and characterization of the PC, emphasizing the in vivo environmental factors that control the PC formation. The effect of PC on the physicochemical properties and thus biological behaviors of nanocarriers was then presented and thoroughly discussed. Finally, we proposed the design strategies available for engineering PC onto nanocarriers to manipulate them with the desired surface properties and achieve the best biomedical outcomes.
Collapse
|
29
|
Mojarad-Jabali S, Mahdinloo S, Farshbaf M, Sarfraz M, Fatahi Y, Atyabi F, Valizadeh H. Transferrin receptor-mediated liposomal drug delivery: recent trends in targeted therapy of cancer. Expert Opin Drug Deliv 2022; 19:685-705. [PMID: 35698794 DOI: 10.1080/17425247.2022.2083106] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Compared to normal cells, malignant cancer cells require more iron for their growth and rapid proliferation, which can be supplied by a high expression level of transferrin receptor (TfR). It is well known that the expression of TfR on the tumor cells is considerably higher than that of normal cells, which makes TfR an attractive target in cancer therapy. AREAS COVERED In this review, the primary focus is on the role of TfR as a valuable tool for cancer-targeted drug delivery, followed by the full coverage of available TfR ligands and their conjugation chemistry to the surface of liposomes. Finally, the most recent studies investigating the potential of TfR-targeted liposomes as promising drug delivery vehicles to different cancer cells are highlighted with emphasis on their improvement possibilities to become a part of future cancer medicines. EXPERT OPINION Liposomes as a valuable class of nanocarriers have gained much attention toward cancer therapy. From all the studies that have exploited the therapeutic and diagnostic potential of TfR on cancer cells, it can be realized that the systematic assessment of TfR ligands applied for liposomal targeted delivery has yet to be entirely accomplished.
Collapse
Affiliation(s)
- Solmaz Mojarad-Jabali
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Student research committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Mahdinloo
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Student research committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Farshbaf
- Student research committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Muhammad Sarfraz
- College of Pharmacy, Al Ain University, Al Ain, United Arab Emirates
| | - Yousef Fatahi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hadi Valizadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
30
|
Wang T, Qin J, Cheng J, Li C, Du J. Intelligent design of polymersomes for antibacterial and anticancer applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1822. [PMID: 35673991 DOI: 10.1002/wnan.1822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 01/25/2023]
Abstract
Polymersomes (or polymer vesicles) have attracted much attention for biomedical applications in recent years because their lumen can be used for drug delivery and their coronas and membrane can be modified with a variety of functional groups. Thus, polymersomes are very suitable for improved antibacterial and anticancer therapy. This review mainly highlighted recent advances in the synthetic protocols and design principles of intelligent antibacterial and anticancer polymersomes. Antibacterial polymersomes are divided into three categories: polymersomes as antibiotic nanocarriers, intrinsically antibacterial polymersomes, and antibacterial polymersomes with supplementary means including photothermal and photodynamic therapy. Similarly, the anticancer polymersomes are divided into two categories: polymersomes-based delivery systems and anticancer polymersomes with supplementary means. In addition, the bilateral relationship between bacteria and cancer is addressed, since more and more evidences show that bacteria may cause cancer or promote cancer progression. Finally, prospective on next-generation antibacterial and anticancer polymersomes are discussed. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Tao Wang
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China
| | - Jinlong Qin
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China.,Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiajing Cheng
- Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Li
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China
| | - Jianzhong Du
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China.,Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
31
|
Nano-Drug Delivery Systems Based on Different Targeting Mechanisms in the Targeted Therapy of Colorectal Cancer. Molecules 2022; 27:molecules27092981. [PMID: 35566331 PMCID: PMC9099628 DOI: 10.3390/molecules27092981] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/23/2022] [Accepted: 05/03/2022] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a usual digestive tract malignancy and the third main cause of cancer death around the world, with a high occurrence rate and mortality rate. Conventional therapies for CRC have certain side effects and restrictions. However, the exciting thing is that with the rapid development of nanotechnology, nanoparticles have gradually become more valuable drug delivery systems than traditional therapies because of their capacity to control drug release and target CRC. This also promotes the application of nano-drug targeted delivery systems in the therapy of CRC. Moreover, to make nanoparticles have a better colon targeting effect, many approaches have been used, including nanoparticles targeting CRC and in response to environmental signals. In this review, we focus on various targeting mechanisms of CRC-targeted nanoparticles and their latest research progress in the last three years, hoping to give researchers some inspiration on the design of CRC-targeted nanoparticles.
Collapse
|
32
|
Wei J, Wu D, Shao Y, Guo B, Jiang J, Chen J, Zhang J, Meng F, Zhong Z. ApoE-mediated systemic nanodelivery of granzyme B and CpG for enhanced glioma immunotherapy. J Control Release 2022; 347:68-77. [PMID: 35513207 DOI: 10.1016/j.jconrel.2022.04.048] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/14/2022] [Accepted: 04/28/2022] [Indexed: 02/07/2023]
Abstract
The response of malignant glioma to immunotherapy remains gloomy due to its discrete immunological environment and poor brain penetration of immunotherapeutic agents. Here, we disclose that ApoE peptide-mediated systemic nanodelivery of granzyme B (GrB) and CpG ODN co-stimulates enhanced immunotherapy of murine malignant LCPN glioma model. ApoE peptide-functionalized polymersomes encapsulating GrB (ApoE-PS-GrB) could effectively penetrate the blood-brain barrier-mimicking endothelial cell monolayer in vitro and further be taken up by LCPN cells, inducing strong immunogenic cell death (ICD). The co-administration of ApoE-PS-GrB and ApoE-PS-CpG in orthotopic LCPN glioma-bearing mice co-stimulated cytokine production, maturation of dendritic cells (DCs), infiltration of cytotoxic T lymphocytes (CTLs) while reduction of regulatory T lymphocytes (Treg) and M2 phenotype macrophages in the tumor microenvironment, leading to greatly delayed tumor progression and significantly prolonged survival time compared with all controls. The ApoE-mediated systemic nanodelivery of GrB and CpG ODN opens a new pathway for potent immunotherapy of malignant glioma.
Collapse
Affiliation(s)
- Jingjing Wei
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Di Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, PR China
| | - Yu Shao
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou 215123, PR China
| | - Beibei Guo
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China
| | - Jingjing Jiang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China
| | - Jian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, PR China; Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, 102206, PR China
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou 215123, PR China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
33
|
Zhang J, Jiang J, Lin S, Cornel EJ, Li C, Du J. Polymersomes: from macromolecular self‐assembly to particle assembly. CHINESE J CHEM 2022. [DOI: 10.1002/cjoc.202200182] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Jiamin Zhang
- Department of Polymeric Materials School of Materials Science and Engineering, Tongji University 4800 Caoan Road Shanghai 201804 China
| | - Jinhui Jiang
- Department of Polymeric Materials School of Materials Science and Engineering, Tongji University 4800 Caoan Road Shanghai 201804 China
| | - Sha Lin
- Department of Polymeric Materials School of Materials Science and Engineering, Tongji University 4800 Caoan Road Shanghai 201804 China
| | - Erik Jan Cornel
- Department of Polymeric Materials School of Materials Science and Engineering, Tongji University 4800 Caoan Road Shanghai 201804 China
| | - Chang Li
- Department of Polymeric Materials School of Materials Science and Engineering, Tongji University 4800 Caoan Road Shanghai 201804 China
| | - Jianzhong Du
- Department of Polymeric Materials School of Materials Science and Engineering, Tongji University 4800 Caoan Road Shanghai 201804 China
- Department of Gynaecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine Tongji University Shanghai 200434 China
| |
Collapse
|
34
|
Nienhaus K, Xue Y, Shang L, Nienhaus GU. Protein adsorption onto nanomaterials engineered for theranostic applications. NANOTECHNOLOGY 2022; 33:262001. [PMID: 35294940 DOI: 10.1088/1361-6528/ac5e6c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/15/2022] [Indexed: 06/14/2023]
Abstract
The key role of biomolecule adsorption onto engineered nanomaterials for therapeutic and diagnostic purposes has been well recognized by the nanobiotechnology community, and our mechanistic understanding of nano-bio interactions has greatly advanced over the past decades. Attention has recently shifted to gaining active control of nano-bio interactions, so as to enhance the efficacy of nanomaterials in biomedical applications. In this review, we summarize progress in this field and outline directions for future development. First, we briefly review fundamental knowledge about the intricate interactions between proteins and nanomaterials, as unraveled by a large number of mechanistic studies. Then, we give a systematic overview of the ways that protein-nanomaterial interactions have been exploited in biomedical applications, including the control of protein adsorption for enhancing the targeting efficiency of nanomedicines, the design of specific protein adsorption layers on the surfaces of nanomaterials for use as drug carriers, and the development of novel nanoparticle array-based sensors based on nano-bio interactions. We will focus on particularly relevant and recent examples within these areas. Finally, we conclude this topical review with an outlook on future developments in this fascinating research field.
Collapse
Affiliation(s)
- Karin Nienhaus
- Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), D-76131 Karlsruhe, Germany
| | - Yumeng Xue
- State Key Laboratory of Solidification Processing, Center for Nano Energy Materials, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Li Shang
- State Key Laboratory of Solidification Processing, Center for Nano Energy Materials, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Gerd Ulrich Nienhaus
- Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), D-76131 Karlsruhe, Germany
- Institute of Nanotechnology, Karlsruhe Institute of Technology (KIT), D-76344 Eggenstein-Leopoldshafen, Germany
- Institute of Biological and Chemical Systems, Karlsruhe Institute of Technology (KIT), D-76344 Eggenstein-Leopoldshafen, Germany
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| |
Collapse
|
35
|
Pramanik A, Xu Z, Shamsuddin SH, Khaled YS, Ingram N, Maisey T, Tomlinson D, Coletta PL, Jayne D, Hughes TA, Tyler AII, Millner PA. Affimer Tagged Cubosomes: Targeting of Carcinoembryonic Antigen Expressing Colorectal Cancer Cells Using In Vitro and In Vivo Models. ACS APPLIED MATERIALS & INTERFACES 2022; 14:11078-11091. [PMID: 35196008 PMCID: PMC9007418 DOI: 10.1021/acsami.1c21655] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/17/2022] [Indexed: 05/10/2023]
Abstract
Nanomedicines, while having been approved for cancer therapy, present many challenges such as low stability, rapid clearance, and nonspecificity leading to off-target toxicity. Cubosomes are porous lyotropic liquid crystalline nanoparticles that have shown great premise as drug delivery vehicles; however, their behavior in vivo is largely underexplored, hindering clinical translation. Here, we have engineered cubosomes based on the space group Im3m that are loaded with copper acetylacetonate as a model drug, and their surfaces are functionalized for the first time with Affimer proteins via copper-free click chemistry to actively target overexpressed carcinoembryonic antigens on LS174T colorectal cancer cells. Unlike nontargeted cubosomes, Affimer tagged cubosomes showed preferential accumulation in cancer cells compared to normal cells not only in vitro (2D monolayer cell culture and 3D spheroid models) but also in vivo in colorectal cancer mouse xenografts, while exhibiting low nonspecific absorption and toxicity in other vital organs. Cancerous spheroids had maximum cell death compared to noncancerous cells upon targeted delivery. Xenografts subjected to targeted drug-loaded cubosomes showed a 5-7-fold higher drug accumulation in the tumor tissue compared to the liver, kidneys, and other vital organs, a significant decrease in tumor growth, and an increased survival rate compared to the nontargeted group. This work encompasses the first thorough preclinical investigation of Affimer targeted cubosomes as a cancer therapeutic.
Collapse
Affiliation(s)
- Arindam Pramanik
- School
of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
- School
of Medicine, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Zexi Xu
- School
of Food Science and Nutrition, University
of Leeds, Leeds LS2 9JT, United Kingdom
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Shazana H. Shamsuddin
- School
of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
- Department
of Pathology, School of Medical Sciences, Universiti Sains Malaysia, George Town 16150, Malaysia
| | - Yazan S. Khaled
- School
of Medicine, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Nicola Ingram
- Leeds Institute
of Medical Research, St James’s University
Hospital, Leeds LS9 7TF, United Kingdom
| | - Thomas Maisey
- School
of Medicine, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Darren Tomlinson
- Biomedical
Health Research Centre, BioScreening Technology Group, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - P. Louise Coletta
- Leeds Institute
of Medical Research, St James’s University
Hospital, Leeds LS9 7TF, United Kingdom
| | - David Jayne
- Leeds Institute
of Medical Research, St James’s University
Hospital, Leeds LS9 7TF, United Kingdom
| | - Thomas A. Hughes
- School
of Medicine, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Arwen I. I. Tyler
- School
of Food Science and Nutrition, University
of Leeds, Leeds LS2 9JT, United Kingdom
| | - Paul A. Millner
- School
of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
36
|
Hernández Becerra E, Quinchia J, Castro C, Orozco J. Light-Triggered Polymersome-Based Anticancer Therapeutics Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:836. [PMID: 35269324 PMCID: PMC8912464 DOI: 10.3390/nano12050836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 01/25/2023]
Abstract
Polymersomes are biomimetic cell membrane-like model structures that are self-assembled stepwise from amphiphilic copolymers. These polymeric (nano)carriers have gained the scientific community's attention due to their biocompatibility, versatility, and higher stability than liposomes. Their tunable properties, such as composition, size, shape, and surface functional groups, extend encapsulation possibilities to either hydrophilic or hydrophobic cargoes (or both) and their site-specific delivery. Besides, polymersomes can disassemble in response to different stimuli, including light, for controlling the "on-demand" release of cargo that may also respond to light as photosensitizers and plasmonic nanostructures. Thus, polymersomes can be spatiotemporally stimulated by light of a wide wavelength range, whose exogenous response may activate light-stimulable moieties, enhance the drug efficacy, decrease side effects, and, thus, be broadly employed in photoinduced therapy. This review describes current light-responsive polymersomes evaluated for anticancer therapy. It includes light-activable moieties' features and polymersomes' composition and release behavior, focusing on recent advances and applications in cancer therapy, current trends, and photosensitive polymersomes' perspectives.
Collapse
Affiliation(s)
- Elisa Hernández Becerra
- Max Planck Tandem Group in Nanobioengineering, Institute of Chemistry, Faculty of Natural and Exact Sciences, University of Antioquia, Complejo Ruta N, Calle 67 No. 52-20, Medellín 050010, Colombia; (E.H.B.); (J.Q.)
| | - Jennifer Quinchia
- Max Planck Tandem Group in Nanobioengineering, Institute of Chemistry, Faculty of Natural and Exact Sciences, University of Antioquia, Complejo Ruta N, Calle 67 No. 52-20, Medellín 050010, Colombia; (E.H.B.); (J.Q.)
| | - Cristina Castro
- Engineering School, Pontificia Bolivariana University, Bloque 11, Cq. 1 No. 70-01, Medellín 050004, Colombia;
| | - Jahir Orozco
- Max Planck Tandem Group in Nanobioengineering, Institute of Chemistry, Faculty of Natural and Exact Sciences, University of Antioquia, Complejo Ruta N, Calle 67 No. 52-20, Medellín 050010, Colombia; (E.H.B.); (J.Q.)
| |
Collapse
|
37
|
Fang H, Sha Y, Yang L, Jiang J, Yin L, Li J, Li B, Klumperman B, Zhong Z, Meng F. Macrophage-Targeted Hydroxychloroquine Nanotherapeutics for Rheumatoid Arthritis Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:8824-8837. [PMID: 35156814 DOI: 10.1021/acsami.1c23429] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with unclear pathogenesis. Hydroxychloroquine (HCQ), despite its moderate anti-RA efficacy, is among the few clinical drugs used for RA therapy. Macrophages reportedly play a vital role in RA. Here, we designed and explored macrophage-targeted HCQ nanotherapeutics based on mannose-functionalized polymersomes (MP-HCQ) for RA therapy. Notably, MP-HCQ exhibited favorable properties of less than 50 nm size, glutathione-accelerated HCQ release, and M1 phenotype macrophage (M1M) targetability, leading to repolarization of macrophages to anti-inflammatory M2 phenotype (M2M), reduced secretion of pro-inflammatory cytokines (IL-6), and upregulation of anti-inflammatory cytokines (IL-10). The therapeutic studies in the zymosan-induced RA (ZIA) mouse model showed marked accumulation of MP-HCQ in the inflammation sites, ameliorated symptoms of RA joints, significantly reduced IL-6, TNF-α, and IL-1β, and increased IL-10 and TGF-β compared with free HCQ. The analyses of RA joints disclosed greatly amplified M2M and declined mature DCs, CD4+ T cells, and CD8+ T cells. In accordance, MP-HCQ significantly reduced the damage of RA joints, cartilages, and bones compared to free HCQ and non-targeted controls. Macrophage-targeted HCQ nanotherapeutics therefore appears as a highly potent treatment for RA.
Collapse
Affiliation(s)
- Hanghang Fang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou 215123, P. R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Yongjie Sha
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou 215123, P. R. China
| | - Liang Yang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou 215123, P. R. China
| | - Jingjing Jiang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou 215123, P. R. China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, P. R. China
| | - Jiaying Li
- Orthopedic Institute, Soochow University, Suzhou 215007, PR China
| | - Bin Li
- Orthopedic Institute, Soochow University, Suzhou 215007, PR China
| | - Bert Klumperman
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland 7602, South Africa
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou 215123, P. R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
38
|
Du B, Jiao Q, Bai Y, Yu M, Pang M, Zhao M, Ma H, Yao H. Glutamine Metabolism-Regulated Nanoparticles to Enhance Chemoimmunotherapy by Increasing Antigen Presentation Efficiency. ACS APPLIED MATERIALS & INTERFACES 2022; 14:8753-8765. [PMID: 35138815 DOI: 10.1021/acsami.1c21417] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although the strategies to induce dendritic cells (DCs) maturation and promote their antigen presentation can stimulate the tumor immune response, the endogenous deficiency and immunosuppression of DCs reduce antigen utilization, which limits antigen presentation efficiency and reduces immunotherapy effectiveness. Here, we report an endogenous stimulus-responsive nanodelivery system (DOX@HFn-MSO@PGZL). On the one hand, doxorubicin (DOX) promoted antigen presentation by DCs after the immunogenic death of tumor cells. On the other hand, l-methionine sulfoximine (MSO) regulated the glutamine metabolism of tumor-associated macrophages (TAMs) to induce a shift toward the M1-type. M1-TAMs synergistically presented antigens with mature DCs and were more frequently produced to destroy the tumor suppressive immune microenvironment, resulting in the alleviation of DCs functional inhibition. Ultimately, the antigen presentation efficiency was improved, completely activating tumor immunity and exhibiting powerful antitumor effects.
Collapse
Affiliation(s)
- Bin Du
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, 100 Science Road, Zhengzhou 450001, China
| | - Qingqing Jiao
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Yimeng Bai
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Min Yu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Mengxue Pang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Mengmeng Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Huizhen Ma
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
| | - Hanchun Yao
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, 100 Science Road, Zhengzhou 450001, China
| |
Collapse
|
39
|
Vyas D, Patel M, Wairkar S. Strategies for active tumor targeting-an update. Eur J Pharmacol 2022; 915:174512. [PMID: 34555395 DOI: 10.1016/j.ejphar.2021.174512] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/03/2021] [Accepted: 09/17/2021] [Indexed: 01/26/2023]
Abstract
A complete cure for cancer is still the holy grail for scientists. The existing treatment of cancer is primarily focused on surgery, radiation and conventional chemotherapy. However, chemotherapeutic agents also affect healthy tissues or organs due to a lack of specificity. While passive targeting is studied for anticancer drugs focused on the enhanced permeability and retention effect, it failed to achieve drug accumulation at the tumor site and desired therapeutic efficacy. This review presents an outline of the current significant targets for active tumor drug delivery systems and provides insight into the direction of active tumor-targeting strategies. For this purpose, a systematic understanding of the physiological factors, tumor microenvironment and its components, overexpressed receptor and associated proteins are covered here. We focused on angiogenesis mediated targeting, receptor-mediated targeting and peptide targeting. This active targeting along with integration with nano delivery systems helps in achieving specific action, thus reducing the associated adverse effects to healthy tissues. Although the tumor-targeting methods and possibilities explored so far seem revolutionary in cancer treatment, in-depth clinical studies data is required for its commercial translation.
Collapse
Affiliation(s)
- Darshan Vyas
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Mital Patel
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India.
| |
Collapse
|
40
|
Ma Z, Foda MF, Zhao Y, Han H. Multifunctional Nanosystems with Enhanced Cellular Uptake for Tumor Therapy. Adv Healthc Mater 2022; 11:e2101703. [PMID: 34626528 DOI: 10.1002/adhm.202101703] [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: 08/17/2021] [Revised: 10/01/2021] [Indexed: 11/10/2022]
Abstract
Rapid development of nanotechnology provides promising strategies in biomedicine, especially in tumor therapy. In particular, the cellular uptake of nanosystems is not only a basic premise to realize various biomedical applications, but also a fatal factor for determining the final therapeutic effect. Thus, a systematic and comprehensive summary is necessary to overview the recent research progress on the improvement of nanosystem cellular uptake for cancer treatment. According to the process of nanosystems entering the body, they can be classified into three categories. The first segment is to enhance the accumulation and permeation of nanosystems to tumor cells through extracellular microenvironment stimulation. The second segment is to improve cellular internalization from extracellular to intracellular via active targeting. The third segment is to enhance the intracellular retention of therapeutics by subcellular localization. The major factors in the delivery can be utilized to develop multifunctional nanosystems for strengthening the tumor therapy. Ultimately, the key challenges and prospective in the emerging research frontier are thoroughly outlined. This review is expected to provide inspiring ideas, promising strategies and potential pathways for developing advanced anticancer nanosystems in clinical practice.
Collapse
Affiliation(s)
- Zhaoyu Ma
- State Key Laboratory of Agricultural Microbiology College of Life Science and Technology Huazhong Agricultural University Wuhan Hubei 430070 P. R. China
- Division of Chemistry and Biological Chemistry School of Physical and Mathematical Sciences Nanyang Technological University 21 Nanyang Link Singapore 637371 Singapore
| | - Mohamed F. Foda
- State Key Laboratory of Agricultural Microbiology College of Life Science and Technology Huazhong Agricultural University Wuhan Hubei 430070 P. R. China
- Department of Biochemistry Faculty of Agriculture Benha University Moshtohor Toukh 13736 Egypt
| | - Yanli Zhao
- State Key Laboratory of Agricultural Microbiology College of Life Science and Technology Huazhong Agricultural University Wuhan Hubei 430070 P. R. China
- Division of Chemistry and Biological Chemistry School of Physical and Mathematical Sciences Nanyang Technological University 21 Nanyang Link Singapore 637371 Singapore
| | - Heyou Han
- State Key Laboratory of Agricultural Microbiology College of Life Science and Technology Huazhong Agricultural University Wuhan Hubei 430070 P. R. China
| |
Collapse
|
41
|
Jiang Y, Jiang Z, Wang M, Ma L. Current understandings and clinical translation of nanomedicines for breast cancer therapy. Adv Drug Deliv Rev 2022; 180:114034. [PMID: 34736986 DOI: 10.1016/j.addr.2021.114034] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most frequently diagnosed cancers that is threatening women's life. Current clinical treatment regimens for breast cancer often involve neoadjuvant and adjuvant systemic therapies, which somewhat are associated with unfavorable features. Also, the heterogeneous nature of breast cancers requires precision medicine that cannot be fulfilled by a single type of systemically administered drug. Taking advantage of the nanocarriers, nanomedicines emerge as promising therapeutic agents for breast cancer that could resolve the defects of drugs and achieve precise drug delivery to almost all sites of primary and metastatic breast tumors (e.g. tumor vasculature, tumor stroma components, breast cancer cells, and some immune cells). Seven nanomedicines as represented by Doxil® have been approved for breast cancer clinical treatment so far. More nanomedicines including both non-targeting and active targeting nanomedicines are being evaluated in the clinical trials. However, we have to realize that the translation of nanomedicines, particularly the active targeting nanomedicines is not as successful as people have expected. This review provides a comprehensive landscape of the nanomedicines for breast cancer treatment, from laboratory investigations to clinical applications. We also highlight the key advances in the understanding of the biological fate and the targeting strategies of breast cancer nanomedicine and the implications to clinical translation.
Collapse
|
42
|
Zhang Z, Cheng X, Jiang H, Gu J, Yin Y, Shen Z, Xu C, Pu Z, Li JB, Xu G. Quantitative proteomic analysis of glycosylated proteins enriched from urine samples with magnetic ConA nanoparticles identifies potential biomarkers for small cell lung cancer. J Pharm Biomed Anal 2021; 206:114352. [PMID: 34509662 DOI: 10.1016/j.jpba.2021.114352] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/21/2021] [Accepted: 08/28/2021] [Indexed: 01/08/2023]
Abstract
Lung cancer has high morbidity and mortality and small cell lung cancer (SCLC) is a highly invasive malignant tumor with a very unfavorable survival rate. Early diagnosis and treatment can result in better prognosis for the SCLC patients but current diagnostic methods are either invasive or incapable for large-scale screen. Therefore, discovering biomarkers for early diagnosis of SCLC is of importance. In this work, we covalently coupled Concanavalin A (ConA) to functionalized magnetic nanoparticles to obtain magnetic ConA-nanoparticles (ConA-NPs) for the enrichment of glycosylated proteins. We then purified glycosylated proteins in 36 urine samples from 9 healthy controls, 9 SCLC patients, 9 lung adenocarcinoma (LUAD) patients, and 9 lung squamous cell carcinoma (LUSC) patients. The purified glycosylated proteins were digested and analyzed by LC-MS/MS for identification and quantification. Among the 398 identified proteins, 20, 15, and 1 glycosylated protein(s), respectively, were upregulated in the urine of SCLC, LUAD, and LUSC patients. Immunoblotting experiments further demonstrated that cathepsin C and transferrin were significantly upregulated in the ConA-NP purified urine of SCLC patients. This work suggests that glycosylated cathepsin C and transferrin might be able to serve as potential biomarkers for the noninvasive diagnosis of SCLC patients.
Collapse
Affiliation(s)
- Zhiyu Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China; Medical School of Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Xinyu Cheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China; Medical School of Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Honglv Jiang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Jingyu Gu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China; Medical School of Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Yunfei Yin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China; Medical School of Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Zhijia Shen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China; Medical School of Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Changgang Xu
- School of Materials Science and Engineering, Xi'an University of Science and Technology, Xi'an 710054, China
| | - Zhongjian Pu
- Department of Oncology, Haian Hospital of Traditional Chinese Medicine, Haian, Jiangsu 226600, China
| | - Jia-Bin Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China.
| | - Guoqiang Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
43
|
The Evolution and Future of Targeted Cancer Therapy: From Nanoparticles, Oncolytic Viruses, and Oncolytic Bacteria to the Treatment of Solid Tumors. NANOMATERIALS 2021; 11:nano11113018. [PMID: 34835785 PMCID: PMC8623458 DOI: 10.3390/nano11113018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 02/07/2023]
Abstract
While many classes of chemotherapeutic agents exist to treat solid tumors, few can generate a lasting response without substantial off-target toxicity despite significant scientific advancements and investments. In this review, the paths of development for nanoparticles, oncolytic viruses, and oncolytic bacteria over the last 20 years of research towards clinical translation and acceptance as novel cancer therapeutics are compared. Novel nanoparticle, oncolytic virus, and oncolytic bacteria therapies all start with a common goal of accomplishing therapeutic drug activity or delivery to a specific site while avoiding off-target effects, with overlapping methodology between all three modalities. Indeed, the degree of overlap is substantial enough that breakthroughs in one therapeutic could have considerable implications on the progression of the other two. Each oncotherapeutic modality has accomplished clinical translation, successfully overcoming the potential pitfalls promising therapeutics face. However, once studies enter clinical trials, the data all but disappears, leaving pre-clinical researchers largely in the dark. Overall, the creativity, flexibility, and innovation of these modalities for solid tumor treatments are greatly encouraging, and usher in a new age of pharmaceutical development.
Collapse
|
44
|
Loh JS, Tan LKS, Lee WL, Ming LC, How CW, Foo JB, Kifli N, Goh BH, Ong YS. Do Lipid-based Nanoparticles Hold Promise for Advancing the Clinical Translation of Anticancer Alkaloids? Cancers (Basel) 2021; 13:5346. [PMID: 34771511 PMCID: PMC8582402 DOI: 10.3390/cancers13215346] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Since the commercialization of morphine in 1826, numerous alkaloids have been isolated and exploited effectively for the betterment of mankind, including cancer treatment. However, the commercialization of alkaloids as anticancer agents has generally been limited by serious side effects due to their lack of specificity to cancer cells, indiscriminate tissue distribution and toxic formulation excipients. Lipid-based nanoparticles represent the most effective drug delivery system concerning clinical translation owing to their unique, appealing characteristics for drug delivery. To the extent of our knowledge, this is the first review to compile in vitro and in vivo evidence of encapsulating anticancer alkaloids in lipid-based nanoparticles. Alkaloids encapsulated in lipid-based nanoparticles have generally displayed enhanced in vitro cytotoxicity and an improved in vivo efficacy and toxicity profile than free alkaloids in various cancers. Encapsulated alkaloids also demonstrated the ability to overcome multidrug resistance in vitro and in vivo. These findings support the broad application of lipid-based nanoparticles to encapsulate anticancer alkaloids and facilitate their clinical translation. The review then discusses several limitations of the studies analyzed, particularly the discrepancies in reporting the pharmacokinetics, biodistribution and toxicity data. Finally, we conclude with examples of clinically successful encapsulated alkaloids that have received regulatory approval and are undergoing clinical evaluation.
Collapse
Affiliation(s)
- Jian Sheng Loh
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Malaysia; (J.S.L.); (C.W.H.)
| | - Li Kar Stella Tan
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor’s University, Jalan Taylors 1, Subang Jaya 47500, Malaysia; (L.K.S.T.); (J.B.F.)
| | - Wai Leng Lee
- School of Science, Monash University Malaysia, Subang Jaya 47500, Malaysia;
| | - Long Chiau Ming
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei; (L.C.M.); (N.K.)
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Malaysia; (J.S.L.); (C.W.H.)
- Health and Well-Being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Subang Jaya 47500, Malaysia
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor’s University, Jalan Taylors 1, Subang Jaya 47500, Malaysia; (L.K.S.T.); (J.B.F.)
- Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health & Medical Sciences, Taylor’s University, Jalan Taylors 1, Subang Jaya 47500, Malaysia
| | - Nurolaini Kifli
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei; (L.C.M.); (N.K.)
| | - Bey Hing Goh
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Malaysia; (J.S.L.); (C.W.H.)
- Biofunctional Molecule Exploratory Research Group (BMEX), School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yong Sze Ong
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Malaysia; (J.S.L.); (C.W.H.)
- Health and Well-Being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Subang Jaya 47500, Malaysia
- Biofunctional Molecule Exploratory Research Group (BMEX), School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Malaysia
| |
Collapse
|
45
|
Zhuo Z, Wang J, Luo Y, Zeng R, Zhang C, Zhou W, Guo K, Wu H, Sha W, Chen H. Targeted extracellular vesicle delivery systems employing superparamagnetic iron oxide nanoparticles. Acta Biomater 2021; 134:13-31. [PMID: 34284151 DOI: 10.1016/j.actbio.2021.07.027] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 02/05/2023]
Abstract
In the past decade, the study of extracellular vesicles (EVs), especially exosomes (50-150 nm) have attracted growing interest in numerous areas of cancer and tissue regeneration due to their unique biological features. A low isolation yield and insufficient targeting abilities limit their therapeutic applicability. Recently, superparamagnetic iron oxide nanoparticles (SPIONs) with magnetic navigation have been exploited to enhance the targeting ability of EVs. To construct targeted EV delivery systems engineered by SPIONs, several groups have pioneered the use of different techniques, such as electroporation, natural incubation, and cell extrusion, to directly internalize SPIONs into EVs. Furthermore, some endogenous ligands, such as transferrins, antibodies, aptamers, and streptavidin, were shown to enable modification of SPIONs, which increases binding with EVs. In this review, we summarized recent advances in targeted EV delivery systems engineered by SPIONs and focused on the key methodological approaches and the current applications of magnetic EVs. This report aims to address the existing challenges and provide comprehensive insights into targeted EV delivery systems. STATEMENT OF SIGNIFICANCE: Targeted extracellular vesicle (EV) delivery systems engineered by superparamagnetic iron oxide nanoparticles (SPIONs) have attracted wide attention and research interest in recent years. Such strategies employ external magnet fields to manipulate SPION-functionalized EVs remotely, aiming to enhance their accumulation and penetration in vivo. Although iron oxide nanoparticle laden EVs are interesting, they are controversial at present, hampering the progress in their clinical application. A thorough integration of these studies is needed for an advanced insight and rational design of targeted EV delivery systems. In this review, we summarize the latest advances in the design strategies of targeted EV delivery systems engineered by SPIONs with a focus on their key methodological approaches, current applications, limitation and future perspectives, which may facilitate the development of natural theranostic nanoplatforms.
Collapse
Affiliation(s)
- Zewei Zhuo
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Jinghua Wang
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yujun Luo
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Shantou University Medical College, Shantou 515041, China
| | - Ruijie Zeng
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Shantou University Medical College, Shantou 515041, China
| | - Chen Zhang
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Weijie Zhou
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Kehang Guo
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Huihuan Wu
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Weihong Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| |
Collapse
|
46
|
Li L, Zhang Q, Li J, Tian Y, Kang Y, Ren G, Liu W, Wang H, Wang B, Yan L, Guo L, Diao H. Targeted Delivery of Doxorubicin Using Transferrin-Conjugated Carbon Dots for Cancer Therapy. ACS APPLIED BIO MATERIALS 2021; 4:7280-7289. [PMID: 35006957 DOI: 10.1021/acsabm.1c00811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A transferrin receptor (TfR)-targeted nanodrug [green fluorescence emission carbon dot (GCD)-polyethylene glycol (PEG)-transferrin (Tf)@doxorubicin (Dox)] for cancer therapy was developed by functionalizing GCDs with PEG, Tf, and Dox. GCDs were synthesized by the one-step hydrothermal method, followed by conjugating PEG and Tf by covalent bonds and loading Dox by electrostatic interactions. The nanodrug exhibits high stability under neutral conditions and effectively releases Dox at pH of 5.5. GCD-PEG-Tf@Dox can be selectively internalized by TfR-overexpressed tumor cells (MCF-7 and K150) via receptor-mediated endocytosis and further release Dox to the nuclei. As a result, GCD-PEG-Tf@Dox exhibits significant lethality to tumor cells (MCF-7 and K150) but greatly reduced toxicity to normal cells [Chinese hamster ovary cell line (CHO)] compared with free Dox. In vivo studies have confirmed that GCD-PEG-Tf@Dox can effectively inhibit tumor proliferation with negligible side effects.
Collapse
Affiliation(s)
- Lihong Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China.,College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.,Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, PR China
| | - Qi Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Jinyao Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yafei Tian
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yu Kang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Guodong Ren
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Wen Liu
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.,Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, PR China
| | - Haojiang Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Bin Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Lili Yan
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Lixia Guo
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Haipeng Diao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China.,College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.,Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, PR China
| |
Collapse
|
47
|
Wei Y, Sun Y, Wei J, Qiu X, Meng F, Storm G, Zhong Z. Selective transferrin coating as a facile strategy to fabricate BBB-permeable and targeted vesicles for potent RNAi therapy of brain metastatic breast cancer in vivo. J Control Release 2021; 337:521-529. [PMID: 34352315 DOI: 10.1016/j.jconrel.2021.07.048] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/31/2021] [Accepted: 07/30/2021] [Indexed: 12/13/2022]
Abstract
Brain metastases are a most disturbing situation for breast cancer patients as there is basically no adequate treatment available. Any potential drug formulation has to be able to cross the blood-brain barrier (BBB) and specific to metastatic brain tumors without causing unacceptable adverse effects. Here, we developed transferrin-functionalized chimeric polymersomes carrying siRNA against polo-like kinase 1 (Tf@TBP-CPs-siPLK1) for treating brain metastatic MDA-MB 231 triple negative breast cancer (TNBC) xenografts in mice. To facilitate the loading of siPLK1, chimaeric polymersomes (CPs) were designed with spermine in the watery core and transferrin-binding peptide (TBP) at the surface, enabling attachment of transferrin after the siRNA loading step and thereby circumventing interference of transferrin with siRNA loading. Tf@TBP-CPs-siPLK1 encapsulating 3.8 wt% siRNA had a mean size of about 50 nm and a neutral zeta potential in phosphate buffer (PB). By virtue of the presence of transferrin, Tf@TBP-CPs demonstrated greatly (ca. 5-fold) enhanced internalization in MDA-MB 231 cells and transcytosis in the endothelial (bEnd.3) monolayer model in vitro as well as markedly improved accumulation in the orthotopically xenografted MDA-MB 231 tumor in the brain in vivo compared with control CPs lacking transferrin, supporting that transferrin mediates efficient BBB penetration and high specificity towards MDA-MB 231 cells. As a result, Tf@TBP-CPs-siPLK1 effectively inhibited tumor progression and prolonged the lifespan of the mice significantly. Selective transferrin coating appears to be a particularly facile strategy to fabricate BBB-permeable and targeted vesicles for potent RNAi therapy of brain metastatic breast cancer.
Collapse
Affiliation(s)
- Yaohua Wei
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Department of Biomaterials Science and Technology, MIRA Institute for Biological Technology and Technical Medicine, University of Twente, PO Box 217, 7500AE Enschede, the Netherlands
| | - Yinping Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Jingjing Wei
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Xinyun Qiu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| | - Gert Storm
- Department of Biomaterials Science and Technology, MIRA Institute for Biological Technology and Technical Medicine, University of Twente, PO Box 217, 7500AE Enschede, the Netherlands; Department of Pharmaceutics, Utrecht University, the Netherlands; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
48
|
Li H, Wang Y, Tang Q, Yin D, Tang C, He E, Zou L, Peng Q. The protein corona and its effects on nanoparticle-based drug delivery systems. Acta Biomater 2021; 129:57-72. [PMID: 34048973 DOI: 10.1016/j.actbio.2021.05.019] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/25/2021] [Accepted: 05/18/2021] [Indexed: 02/04/2023]
Abstract
In most cases, once nanoparticles (NPs) enter the blood, their surface is covered by biological molecules, especially proteins, forming a so-called protein corona (PC). As a result, what the cells of the body "see" is not the NPs as formulated by the chemists, but the PC. In this way, the PC can influence the effects of the NPs and even mask the desired effects of the NP components. While this can argue for trying to inhibit protein-nanomaterial interactions, encapsulating NPs in an endogenous PC may increase their clinical usefulness. In this review, we briefly introduce the concept of the PC, its formation and its effects on the behavior of NPs. We also discuss how to reduce the formation of PCs or exploit them to enhance NP functions. Studying the interactions between proteins and NPs will provide insights into their clinical activity in health and disease. STATEMENT OF SIGNIFICANCE: The formation of protein corona (PC) will affect the operation of nanoparticles (NPs) in vivo. Since there are many proteins in the blood, it is impossible to completely overcome the formation of PC. Therefore, the use of PCs to deliver drug is the best choice. De-opsonins adsorbed on NPs can reduce macrophage phagocytosis and cytotoxicity of NPs, and prolong their circulation in blood. Albumin, apolipoprotein and transferrin are typical de-opsonins. In present review, we mainly discuss how to optimize the delivery of nanoparticles through the formation of albumin corona, transferrin corona and apolipoprotein corona in vivo or in vitro.
Collapse
Affiliation(s)
- Hanmei Li
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China
| | - Yao Wang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Qi Tang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Dan Yin
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Chuane Tang
- School of Mechanical Engineering, Chengdu university, Chengdu 610106, China
| | - En He
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
49
|
Dimitriou P, Li J, Tornillo G, McCloy T, Barrow D. Droplet Microfluidics for Tumor Drug-Related Studies and Programmable Artificial Cells. GLOBAL CHALLENGES (HOBOKEN, NJ) 2021; 5:2000123. [PMID: 34267927 PMCID: PMC8272004 DOI: 10.1002/gch2.202000123] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/19/2021] [Indexed: 05/11/2023]
Abstract
Anticancer drug development is a crucial step toward cancer treatment, that requires realistic predictions of malignant tissue development and sophisticated drug delivery. Tumors often acquire drug resistance and drug efficacy, hence cannot be accurately predicted in 2D tumor cell cultures. On the other hand, 3D cultures, including multicellular tumor spheroids (MCTSs), mimic the in vivo cellular arrangement and provide robust platforms for drug testing when grown in hydrogels with characteristics similar to the living body. Microparticles and liposomes are considered smart drug delivery vehicles, are able to target cancerous tissue, and can release entrapped drugs on demand. Microfluidics serve as a high-throughput tool for reproducible, flexible, and automated production of droplet-based microscale constructs, tailored to the desired final application. In this review, it is described how natural hydrogels in combination with droplet microfluidics can generate MCTSs, and the use of microfluidics to produce tumor targeting microparticles and liposomes. One of the highlights of the review documents the use of the bottom-up construction methodologies of synthetic biology for the formation of artificial cellular assemblies, which may additionally incorporate both target cancer cells and prospective drug candidates, as an integrated "droplet incubator" drug assay platform.
Collapse
Affiliation(s)
- Pantelitsa Dimitriou
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - Jin Li
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - Giusy Tornillo
- Hadyn Ellis BuildingCardiff UniversityMaindy RoadCardiffCF24 4HQUK
| | - Thomas McCloy
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - David Barrow
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| |
Collapse
|
50
|
Novel PVA-Based Microspheres Co-Loaded with Photothermal Transforming Agent and Chemotherapeutic for Colorectal Cancer Treatment. Pharmaceutics 2021; 13:pharmaceutics13070984. [PMID: 34209684 PMCID: PMC8309159 DOI: 10.3390/pharmaceutics13070984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/06/2021] [Accepted: 06/17/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND We previously designed an electrospinning chitosan (CS) nanofiber-based carrier, using polyvinyl alcohol (PVA) as an adjuvant to deliver doxorubicin (DOX) and MoS2 nanosheets for postoperative tumor re-occurrence inhibition. However, owing to that the nanofibrous mat is un-injectable, this composite nanofiber is far from being clinically applicable. MATERIALS AND METHODS Via modulating the electrospray parameters, polyvinyl alcohol (PVA) beads string doped with DOX and MoS2 (PVA/MoS2/DOX microspheres) were prepared, which were further crosslinked with glutaraldehyde to obtain the water-stability. RESULTS Under the 808-nm laser irradiation, MoS2 nanosheets rendered the prepared PVA/MoS2/DOX microspheres an excellent light-to-heat conversion performance with η of 23.2%. Besides, the heat generated by near-infrared laser irradiation can improve the effect of chemotherapy by promoting the release rate of DOX. HT29 cell and tumor-bearing nude mice were used to systematically study the combined tumor treatment efficiency of composite nanospheres. CONCLUSION PVA/MoS2/DOX nanospheres have excellent photothermal effect and chemotherapy effect, which can completely suppress the tumor recurrence. Therefore, the PVA/MoS2/DOX nanospheres are anticipated to find potential applications in the treatment of local colorectal cancer.
Collapse
|