1
|
Huang T, Huang W, Huang G, Wei X, Huang Y, Liu T, Liu Y, Ni W, Che C. Pincer-Type Pt(II)-NHC Antibody-Drug Conjugate for HER-2-Targeted Chemoimmunotherapy. Adv Healthc Mater 2025; 14:e2403449. [PMID: 39950551 PMCID: PMC11973945 DOI: 10.1002/adhm.202403449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/28/2025] [Indexed: 04/08/2025]
Abstract
Platinum-based chemotherapy drugs play an indispensable role in clinical cancer treatment, but exhibit considerable side effects due to their non-specific mechanism of killing cancer cells and normal cells. In this regard, the use of antibodies conjugated to anti-cancer platinum complexes will enable better differentiation of cancer cells from normal cells. Here, six pincer-platinum(II) NHC (N-heterocyclic carbene) complexes are reported, one of which has an amino group on the N-alkyl group of the NHC ligand. This platinum(II) complex is used as the payload for platinum(II)-based antibody-drug conjugate (ADC) targeting human epidermal growth factor receptor 2 (HER-2). Notably, this ADC can specifically bind to the HER-2 antigen, distinguish target cells from non-target cells, and exhibit good anti-tumor activity in vitro and in vivo.
Collapse
Affiliation(s)
- Tao Huang
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Wan‐Qiong Huang
- Department of PathologyCancer Hospital of Shantou University Medical CollegeShantouGuangdong515041P. R. China
| | - Gui‐Feng Huang
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Xiao‐Long Wei
- Department of PathologyCancer Hospital of Shantou University Medical CollegeShantouGuangdong515041P. R. China
| | - Yong‐Liang Huang
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
| | - Tao Liu
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Yungen Liu
- Department of ChemistrySouthern University of Science and TechnologyShenzhenGuangdong518055P. R. China
| | - Wen‐Xiu Ni
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Chi‐Ming Che
- State Key Laboratory of Synthetic Chemistry and Department of ChemistryThe University of Hong KongHong KongP. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology LimitedHong Kong Science and Technology ParksUnits 1503–1511, 15/F, Building 17W, New TerritoriesHong KongP. R. China
| |
Collapse
|
2
|
Guo T, Hayat MA, Hu J. Ferritin nanoparticles: new strategies for the diagnosis and treatment of central nervous system diseases. Biomed Mater 2025; 20:022005. [PMID: 39820046 DOI: 10.1088/1748-605x/adab5a] [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: 10/23/2024] [Accepted: 01/16/2025] [Indexed: 01/19/2025]
Abstract
Ferritin nanoparticles, which can penetrate the blood-brain barrier (BBB), have gained significant research interest for the diagnosis and treatment of central nervous system (CNS) diseases, including gliomas, Alzheimer's disease, and brain metastases. In recent years, ferritin has been proved as a candidate to cross the BBB using receptor-mediated transport (RMT) mechanism through transferrin receptor 1 (TfR1) which is overexpressed in the cells of the BBB. Various types of cargo molecules, including therapeutics, imaging agents, nucleic acids, and metal nanoparticles, have been incorporated into ferritin nanocages for the diagnosis and treatment of CNS diseases. In particular, low immunogenicity of ferritin implies safety for its usage in clinical practices, and high biocompatibility add to the perspectives of its applications. Furthermore, contemporary strides in molecular biology have enabled some alteration in the configuration of the ferritin outer layers and surface characters so as to enhance the drug encapsulation capacity and conjugation affinity. Such modifications not only enhance the property of ferritin in crossing the BBB, but also enhance its efficacy when applied to CNS diseases. In summary, ferritin, as a drug delivery system, shows great potential for the treatment and diagnosis of CNS diseases.
Collapse
Affiliation(s)
- Tao Guo
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, People's Republic of China
| | - Muhammad Abid Hayat
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, People's Republic of China
| | - Jiabo Hu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, People's Republic of China
- Zhenjiang Blood Center, Zhenjiang, Jiangsu 212013, People's Republic of China
| |
Collapse
|
3
|
Darmadi D, Aminov Z, Hjazi A, R R, Kazmi SW, Mustafa YF, Hosseen B, Sharma A, Alubiady MHS, Al-Abdeen SHZ. Investigation of the regulation of EGF signaling by miRNAs, delving into the underlying mechanism and signaling pathways in cancer. Exp Cell Res 2024; 442:114267. [PMID: 39313176 DOI: 10.1016/j.yexcr.2024.114267] [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: 07/20/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 09/25/2024]
Abstract
The EGF receptors (EGFRs) signaling pathway is essential for tumorigenesis and progression of cancer. Emerging evidence suggests that miRNAs are essential regulators of EGF signaling, influencing various pathway components and tumor behavior. This article discusses the underlying mechanisms and clinical implications of miRNA-mediated regulation of EGF signaling in cancer. miRNAs utilize multiple mechanisms to exert their regulatory effects on EGF signaling. They can target EGF ligands, including EGF and TGF-directly, inhibiting their expression and secretion. In addition, miRNAs can modulate EGF signaling indirectly by targeting EGF receptors, downstream signaling molecules, and transcription factors implicated in regulating the EGF pathway. These miRNAs can disrupt the delicate equilibrium of EGF signaling, resulting in aberrant activation and fostering tumor cell proliferation, survival, angiogenesis, and metastasis. The dysregulation of the expression of specific miRNAs has been linked to clinical outcomes in numerous types of cancer. Specific profiles of miRNA expression have been identified as prognostic markers, reflecting tumor characteristics, invasiveness, metastatic potential, and therapeutic response. These miRNAs can serve as potential therapeutic targets for interventions that modulate EGF signaling and improve patient outcomes. Understanding the intricate relationship between miRNAs and EGF signaling in cancer can transform cancer diagnosis, prognosis, and treatment. The identification of specific miRNAs involved in the regulation of the EGF pathway opens the door to the development of targeted therapies and personalized medicine approaches. In addition, miRNA-based interventions promise to overcome therapeutic resistance and improve the efficacy of existing treatments. miRNAs are crucial regulators of EGF signaling in cancer, affecting tumor behavior and clinical outcomes. Further research is required to decipher the complex network of miRNA-mediated EGF signaling regulation and translate these findings into clinically applicable strategies for enhanced cancer treatment.
Collapse
Affiliation(s)
- Darmadi Darmadi
- Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia.
| | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan.
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia.
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Syeda Wajida Kazmi
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, 140307, Punjab, India.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq.
| | - Beneen Hosseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq; Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq.
| | - Abhishek Sharma
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | | | | |
Collapse
|
4
|
Shatz-Binder W, Azumaya CM, Leonard B, Vuong I, Sudhamsu J, Rohou A, Liu P, Sandoval W, Bol K, Izadi S, Holder PG, Blanchette C, Perozzo R, Kelley RF, Kalia Y. Adapting Ferritin, a Naturally Occurring Protein Cage, to Modulate Intrinsic Agonism of OX40. Bioconjug Chem 2024; 35:593-603. [PMID: 38592684 PMCID: PMC11099885 DOI: 10.1021/acs.bioconjchem.4c00020] [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: 01/19/2024] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/10/2024]
Abstract
Ferritin is a multivalent, self-assembling protein scaffold found in most human cell types, in addition to being present in invertebrates, higher plants, fungi, and bacteria, that offers an attractive alternative to polymer-based drug delivery systems (DDS). In this study, the utility of the ferritin cage as a DDS was demonstrated within the context of T cell agonism for tumor killing. Members of the tumor necrosis factor receptor superfamily (TNFRSF) are attractive targets for the development of anticancer therapeutics. These receptors are endogenously activated by trimeric ligands that occur in transmembrane or soluble forms, and oligomerization and cell-surface anchoring have been shown to be essential aspects of the targeted agonism of this receptor class. Here, we demonstrated that the ferritin cage could be easily tailored for multivalent display of anti-OX40 antibody fragments on its surface and determined that these arrays are capable of pathway activation through cell-surface clustering. Together, these results confirm the utility, versatility, and developability of ferritin as a DDS.
Collapse
Affiliation(s)
- Whitney Shatz-Binder
- Protein
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
- Pharmaceutical
Sciences, University of Geneva, Geneva 1211, Switzerland
| | - Caleigh M. Azumaya
- Structural
Biology, Genentech Inc., South San Francisco, California 94080, United States
| | - Brandon Leonard
- Antibody
Engineering, Genentech Inc., South San Francisco, California 94080, United States
| | - Ivan Vuong
- Protein
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S Ellis Ave, Chicago, Illinois 60637, United States
| | - Jawahar Sudhamsu
- Structural
Biology, Genentech Inc., South San Francisco, California 94080, United States
| | - Alexis Rohou
- Structural
Biology, Genentech Inc., South San Francisco, California 94080, United States
| | - Peter Liu
- Microchemistry,
Proteomics and Lipidomics, Genentech Inc., South San Francisco, California 94080, United States
| | - Wendy Sandoval
- Microchemistry,
Proteomics and Lipidomics, Genentech Inc., South San Francisco, California 94080, United States
| | - Karenna Bol
- Pharmaceutical
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
- Business
and Program Management, Genentech Inc., South San Francisco, California 94080, United States
| | - Saeed Izadi
- Pharmaceutical
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
| | - Patrick G. Holder
- Protein
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
| | - Craig Blanchette
- Protein
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
| | - Remo Perozzo
- Pharmaceutical
Sciences, University of Geneva, Geneva 1211, Switzerland
| | - Robert F. Kelley
- Pharmaceutical
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
| | - Yogeshvar Kalia
- Pharmaceutical
Sciences, University of Geneva, Geneva 1211, Switzerland
| |
Collapse
|
5
|
Liu L, Xue W. Thalidomide suppresses migration and invasion of colorectal cancer cells by inhibiting HOXB7-mediated activation of the Wnt/β-catenin signaling pathway. Chem Biol Drug Des 2024; 103:e14434. [PMID: 38230780 DOI: 10.1111/cbdd.14434] [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: 09/07/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024]
Abstract
Heaps of studies have verified the effects of thalidomide (THA) on colorectal cancer (CRC). Howbeit, the corresponding mechanism awaits illustration, which is the foothold of this study. Following the treatment of 0, 1.94, 7.75, or 19.36 μM THA, CRC cell viability, apoptosis, migration, and invasion were evaluated by methyl tetrazolium, flow cytometry, wound-healing, and transwell assays. Homeobox B7 (HOXB7) expression in CRC was analyzed and detected by bioinformatics analysis, quantitative real-time PCR or western blot. After the corresponding transfection or treatment with inhibitor of catenin-responsive transcription-3 (iCRT-3), abovementioned CRC cell biological behaviors as well as expression levels of HOXB7 and β-catenin were evaluated. 7.75 and 19.36 μM THA dwindled CRC cell viability, migration, and invasion, and facilitated apoptosis. HOXB7 upregulation was detected in CRC cells, which promoted the viability, migration, invasion, and β-catenin expression, and weakened the apoptosis of CRC cells. Also, HOXB7 upregulation counteracted the effects of THA on CRC cells. iCRT-3 restrained β-catenin expression, viability, migration, and invasion, whereas promoting the apoptosis of CRC cells. In addition, iCRT-3 antagonized the effects of overexpressed HOXB7 on CRC cells. THA inhibits the migration and invasion of CRC cells, which is achieved by suppressing HOXB7-mediated activation of Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Liyang Liu
- Department of Anoretal, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wusong Xue
- Department of Anoretal, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Bagheri M, Zandieh MA, Daryab M, Samaei SS, Gholami S, Rahmanian P, Dezfulian S, Eary M, Rezaee A, Rajabi R, Khorrami R, Salimimoghadam S, Hu P, Rashidi M, Ardakan AK, Ertas YN, Hushmandi K. Nanostructures for site-specific delivery of oxaliplatin cancer therapy: Versatile nanoplatforms in synergistic cancer therapy. Transl Oncol 2024; 39:101838. [PMID: 38016356 DOI: 10.1016/j.tranon.2023.101838] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/24/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023] Open
Abstract
As a clinically approved treatment strategy, chemotherapy-mediated tumor suppression has been compromised, and in spite of introducing various kinds of anticancer drugs, cancer eradication with chemotherapy is still impossible. Chemotherapy drugs have been beneficial in improving the prognosis of cancer patients, but after resistance emerged, their potential disappeared. Oxaliplatin (OXA) efficacy in tumor suppression has been compromised by resistance. Due to the dysregulation of pathways and mechanisms in OXA resistance, it is suggested to develop novel strategies for overcoming drug resistance. The targeted delivery of OXA by nanostructures is described here. The targeted delivery of OXA in cancer can be mediated by polymeric, metal, lipid and carbon nanostructures. The advantageous of these nanocarriers is that they enhance the accumulation of OXA in tumor and promote its cytotoxicity. Moreover, (nano)platforms mediate the co-delivery of OXA with drugs and genes in synergistic cancer therapy, overcoming OXA resistance and improving insights in cancer patient treatment in the future. Moreover, smart nanostructures, including pH-, redox-, light-, and thermo-sensitive nanostructures, have been designed for OXA delivery and cancer therapy. The application of nanoparticle-mediated phototherapy can increase OXA's potential in cancer suppression. All of these subjects and their clinical implications are discussed in the current review.
Collapse
Affiliation(s)
- Mohsen Bagheri
- Radiology Resident, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mahshid Daryab
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Setareh Samaei
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sarah Gholami
- Young Researcher and Elite Club, Babol Branch, Islamic Azad University, Babol, Iran
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sadaf Dezfulian
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mahsa Eary
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Peng Hu
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Alireza Khodaei Ardakan
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
7
|
Pan Q, Fan X, Xie L, Wu D, Liu R, Gao W, Luo K, He B, Pu Y. Nano-enabled colorectal cancer therapy. J Control Release 2023; 362:548-564. [PMID: 37683732 DOI: 10.1016/j.jconrel.2023.09.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
Colorectal cancer (CRC), one of the most common and deadliest diseases worldwide, poses a great health threat and social burden. The clinical treatments of CRC encompassing surgery, chemotherapy, and radiotherapy are challenged with toxicity, therapy resistance, and recurrence. In the past two decades, targeted therapy and immunotherapy have greatly improved the therapeutic benefits of CRC patients but they still suffer from drug resistance and low response rates. Very recently, gut microbiota regulation has exhibited a great potential in preventing and treating CRC, as well as in modulating the efficacy and toxicity of chemotherapy and immunotherapy. In this review, we provide a cutting-edge summary of nanomedicine-based treatment in colorectal cancer, highlighting the recent progress of oral and systemic tumor-targeting and/or tumor-activatable drug delivery systems as well as novel therapeutic strategies against CRC, including nano-sensitizing immunotherapy, anti-inflammation, gut microbiota modulation therapy, etc. Finally, the recent endeavors to address therapy resistance, metastasis, and recurrence in CRC were discussed. We hope this review could offer insight into the design and development of nanomedicines for CRC and beyond.
Collapse
Affiliation(s)
- Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Xi Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Li Xie
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Di Wu
- Meat Processing Key Laboratory of Sichuan Province, School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Rong Liu
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China.
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
8
|
Liu Z, Feng Z, Chen M, Zhan J, Wu R, Shi Y, Xue Y, Liu R, Zhu JJ, Zhang J. An orthogonally activatable CRISPR-Cas13d nanoprodrug to reverse chemoresistance for enhanced chemo-photodynamic therapy. Chem Sci 2023; 14:4102-4113. [PMID: 37063792 PMCID: PMC10094006 DOI: 10.1039/d3sc00020f] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Orthogonal therapy that combines CRISPR-based gene editing and prodrug-based chemotherapy is a promising approach to combat multidrug-resistant cancer. However, its potency to precisely regulate different therapeutic modalities in vivo is limited due to the lack of an integrated platform with high spatiotemporal resolution. Taking advantage of CRISPR technology, a Pt(iv)-based prodrug and orthogonal emissive upconversion nanoparticles (UCNPs), we herein rationally designed the first logic-gated CRISPR-Cas13d-based nanoprodrug for orthogonal photomodulation of gene editing and prodrug release for enhanced cancer therapy. The nanoprodrug (URL) was constructed by encapsulating a green light-activatable Pt(iv) prodrug and UV light-activatable Cas13d gene editing tool into UCNPs. We demonstrated that URL maintained excellent orthogonal emission behaviors under 808 and 980 nm excitations, allowing wavelength-selective photoactivation of Cas13d and the prodrug for downregulation of the resistance-related gene and induction of chemo-photodynamic therapy, respectively. Moreover, the photomodulation superiority of URL for overcoming drug resistance was highlighted by integrating it with a Boolean logic gate for programmable modulation of multiple cell behaviors. Importantly, in vivo studies demonstrated that URL can promote Pt(iv) prodrug activation and ROS generation and massively induce on-target drug accumulation by Cas13d-mediated drug resistance attenuation, delivering an ultimate chemo-photodynamic therapeutic performance in efficiently eradicating primary tumors and preventing further liver metastasis. Collectively, our results suggest that URL expands the Cas13d-based genome editing toolbox into prodrug nanomedicine and accelerates the discovery of new orthogonal therapeutic approaches.
Collapse
Affiliation(s)
- Zheng Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Zhiyuan Feng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Mohan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Jiayin Zhan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Rong Wu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Yang Shi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Yunsheng Xue
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University Xuzhou 221004 China
| | - Ran Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Jingjing Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| |
Collapse
|
9
|
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
|
10
|
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: 26] [Impact Index Per Article: 8.7] [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
|
11
|
Wu SY, Wu FG, Chen X. Antibody-Incorporated Nanomedicines for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109210. [PMID: 35142395 DOI: 10.1002/adma.202109210] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/06/2022] [Indexed: 06/14/2023]
Abstract
Antibody-based cancer therapy, one of the most significant therapeutic strategies, has achieved considerable success and progress over the past decades. Nevertheless, obstacles including limited tumor penetration, short circulation half-lives, undesired immunogenicity, and off-target side effects remain to be overcome for the antibody-based cancer treatment. Owing to the rapid development of nanotechnology, antibody-containing nanomedicines that have been extensively explored to overcome these obstacles have already demonstrated enhanced anticancer efficacy and clinical translation potential. This review intends to offer an overview of the advancements of antibody-incorporated nanoparticulate systems in cancer treatment, together with the nontrivial challenges faced by these next-generation nanomedicines. Diverse strategies of antibody immobilization, formats of antibodies, types of cancer-associated antigens, and anticancer mechanisms of antibody-containing nanomedicines are provided and discussed in this review, with an emphasis on the latest applications. The current limitations and future research directions on antibody-containing nanomedicines are also discussed from different perspectives to provide new insights into the construction of anticancer nanomedicines.
Collapse
Affiliation(s)
- Shun-Yu Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119077, Singapore
| |
Collapse
|
12
|
Edwardson TGW, Levasseur MD, Tetter S, Steinauer A, Hori M, Hilvert D. Protein Cages: From Fundamentals to Advanced Applications. Chem Rev 2022; 122:9145-9197. [PMID: 35394752 DOI: 10.1021/acs.chemrev.1c00877] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proteins that self-assemble into polyhedral shell-like structures are useful molecular containers both in nature and in the laboratory. Here we review efforts to repurpose diverse protein cages, including viral capsids, ferritins, bacterial microcompartments, and designed capsules, as vaccines, drug delivery vehicles, targeted imaging agents, nanoreactors, templates for controlled materials synthesis, building blocks for higher-order architectures, and more. A deep understanding of the principles underlying the construction, function, and evolution of natural systems has been key to tailoring selective cargo encapsulation and interactions with both biological systems and synthetic materials through protein engineering and directed evolution. The ability to adapt and design increasingly sophisticated capsid structures and functions stands to benefit the fields of catalysis, materials science, and medicine.
Collapse
Affiliation(s)
| | | | - Stephan Tetter
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Angela Steinauer
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Mao Hori
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Donald Hilvert
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
13
|
Ji P, Wang X, Yin J, Mou Y, Huang H, Ren Z. Selective delivery of curcumin to breast cancer cells by self-targeting apoferritin nanocages with pH-responsive and low toxicity. Drug Deliv 2022; 29:986-996. [PMID: 35363115 PMCID: PMC8979518 DOI: 10.1080/10717544.2022.2056662] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Breast cancer is prevalent and diverse with significantly high incidence and mortality rates. Curcumin (Cur), a polyphenol component of turmeric, has been widely recognized as having strong anti-breast cancer activity. However, its anti-cancer efficiency is largely impaired by some of its concomitant negative properties, including its poor solubility, low cellular uptake, and severe reported side effects. Hence, the necessity arises to develop a novel low-toxic and high-efficiency targeting drug delivery system (DDS). In this study, we developed a pH-sensitive tumor self-targeting DDS (Cur@HFn) based on self-assembled HFn loaded with Cur, in which Cur was encapsulated into HFn cavity by using a disassembly/reassembly strategy, and the Cur@HFn was characterized by ultraviolet–visible (UV–vis), dynamic light scattering (DLS), and transmission electron microscope (TEM). A variety of breast cancer cell models were built to evaluate cytotoxicity, apoptosis, targeting properties, and uptake mechanism of the Cur@HFn. The pharmacodynamics was also evaluated in tumor (4T1) bearing mice after intravenous injection. In vitro release experiments showed that Cur@HFn is pH sensitive and shows sustained drug release under slightly acidic conditions. Compared with Cur, Cur@HFn has stronger cytotoxicity, cellular uptake, and targeting performance. Our study supported that Cur@HFn has a higher in vivo therapeutic effect and lower systemic toxicity. The safety evaluation results indicated that Cur@HFn has no hematotoxicity, hepatotoxicity, and nephrotoxicity. The findings of the present study showed that the Cur@HFn has been successfully prepared and has potential application value in the treatment of breast cancer.
Collapse
Affiliation(s)
- Peng Ji
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou, PR China
| | - Xianglong Wang
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou, PR China
| | - Jiabing Yin
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou, PR China
| | - Yi Mou
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou, PR China
| | - Haiqin Huang
- School of Pharmacy, Nantong University, Nantong, PR China
| | - Zhenkun Ren
- The Third Hospital Affiliated of Jinzhou Medical University, Jinzhou, PR China
| |
Collapse
|
14
|
Franco Machado J, Morais TS. Are smart delivery systems the solution to overcome the lack of selectivity of current metallodrugs in cancer therapy? Dalton Trans 2022; 51:2593-2609. [PMID: 35106525 DOI: 10.1039/d1dt04079k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Chemotherapeutic metallodrugs such as cisplatin and its derivatives are among the most widely applied anticancer treatments worldwide. Despite their clinical success, patients suffer from severe adverse effects while subjected to treatment due to platinum's low selectivity for tumour over healthy tissues. Additionally, intrinsic or acquired resistance to metallodrugs, as well as their inability to reach cancer metastases, often results in therapeutic failure. The evident need for highly efficient and specific treatments has driven the scientific community to research novel ways to surpass the stated limitations. Within this scenario, a rising number of smart drug delivery systems have been lately reported to target primary cancers or metastases, where the metallodrugs are released in a controlled and selective way triggered by specific tumour-related stimuli, thus suggesting a viable and attractive therapeutic approach. Herein, we discuss the main efforts undertaken in the past few years towards the smart delivery of metal-based drugs and drug candidates to tumour sites, particularly focusing on the pH- and/or redox-responsive targeted delivery of platinum and ruthenium anticancer complexes.
Collapse
Affiliation(s)
- João Franco Machado
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal.
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal
| | - Tânia S Morais
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal.
| |
Collapse
|
15
|
Kuruppu AI, Turyanska L, Bradshaw TD, Manickam S, Galhena BP, Paranagama P, De Silva R. Apoferritin and Dps as drug delivery vehicles: Some selected examples in oncology. Biochim Biophys Acta Gen Subj 2022; 1866:130067. [PMID: 34896255 DOI: 10.1016/j.bbagen.2021.130067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/27/2021] [Accepted: 12/02/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND The ideal nanoparticle should be able to encapsulate either pharmaceutical agents or imaging probes so that it could treat or image clinical tumours by targeting the cancer site efficiently. Further, it would be an added advantage if it demonstrates: small size, built in targeting, biocompatibility and biodegradability. Ferritin, which is an endogenous self-assembling protein, stores iron and plays a role in iron homeostasis. When iron atoms are removed apoferritin (AFt) is formed which consists of a hollow shell where it can be used to load guest molecules. Due to its unique architecture, AFt has been investigated as a versatile carrier for tumour theranostic applications. DNA-binding protein from starved cells (Dps), which also belongs to the ferritin family, is a protein found only in prokaryotes. It is used to store iron and protect chromosomes from oxidative damage; because of its architecture, Dps could also be used as a delivery vehicle. CONCLUSIONS Both these nano particles are promising in the field of oncology, especially due to their stability, solubility and biocompatibility features. Further their exterior surface can be modified for better tumour-targeting ability. More studies, are warranted to determine the immunogenicity, biodistribution, and clearance from the body. GENERAL PERSPECTIVE This review discusses a few selected examples of the remarkable in vitro and in vivo studies that have been carried out in the recent past with the use of AFt and Dps in targeting and delivery of various pharmaceutical agents, natural products and imaging probes in the field of oncology.
Collapse
Affiliation(s)
- Anchala I Kuruppu
- Institute for Combinatorial Advanced Research & Education, General Sir John Kotelawala Defence University, Sri Lanka.
| | | | | | - Sivakumar Manickam
- Petroleum and Chemical Engineering, Faculty of Engineering, Universiti Teknologi Brunei, Brunei Darussalam
| | - Bandula Prasanna Galhena
- Department Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Kelaniya, Sri Lanka
| | - Priyani Paranagama
- Department of Chemistry, Faculty of Science, University of Kelaniya, Sri Lanka; Institute of Indigenous Medicine, University of Colombo, Sri Lanka
| | - Ranil De Silva
- Institute for Combinatorial Advanced Research & Education, General Sir John Kotelawala Defence University, Sri Lanka
| |
Collapse
|
16
|
Mainini F, Bonizzi A, Sevieri M, Sitia L, Truffi M, Corsi F, Mazzucchelli S. Protein-Based Nanoparticles for the Imaging and Treatment of Solid Tumors: The Case of Ferritin Nanocages, a Narrative Review. Pharmaceutics 2021; 13:2000. [PMID: 34959283 PMCID: PMC8708614 DOI: 10.3390/pharmaceutics13122000] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022] Open
Abstract
Protein nanocages have been studied extensively, due to their unique architecture, exceptional biocompatibility and highly customization capabilities. In particular, ferritin nanocages (FNs) have been employed for the delivery of a vast array of molecules, ranging from chemotherapeutics to imaging agents, among others. One of the main favorable characteristics of FNs is their intrinsic targeting efficiency toward the Transferrin Receptor 1, which is overexpressed in many tumors. Furthermore, genetic manipulation can be employed to introduce novel variants that are able to improve the loading capacity, targeting capabilities and bio-availability of this versatile drug delivery system. In this review, we discuss the main characteristics of FN and the most recent applications of this promising nanotechnology in the field of oncology with a particular emphasis on the imaging and treatment of solid tumors.
Collapse
Affiliation(s)
- Francesco Mainini
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Arianna Bonizzi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Marta Sevieri
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Leopoldo Sitia
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Fabio Corsi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Serena Mazzucchelli
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| |
Collapse
|
17
|
Nanomedicines functionalized with anti-EGFR ligands for active targeting in cancer therapy: Biological strategy, design and quality control. Int J Pharm 2021; 605:120795. [PMID: 34119579 DOI: 10.1016/j.ijpharm.2021.120795] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
Recently, active targeting using nanocarriers with biological ligands has emerged as a novel strategy for improving the delivery of therapeutic and/or imaging agents to tumor cells. The presence of active targeting moieties on the surface of nanomedicines has been shown to play an important role in enhancing their accumulation in tumoral cells and tissues versus healthy ones. This property not only helps to increase the therapeutic index but also to minimize possible side effects of the designed nanocarriers. Since the overexpression of epidermal growth factor receptors (EGFR) is a common occurrence linked to the progression of a broad variety of cancers, the potential application of anti-EGFR immunotherapy and EGFR-targeting ligands in active targeting nanomedicines is getting increasing attention. Henceforth, the EGFR-targeted nanomedicines were extensively studied in vitro and in vivo but exhibited both satisfactory and disappointing results, depending on used protocols. This review is designed to give an overview of a variety of EGFR-targeting ligands available for nanomedicines, how to conjugate them onto the surface of nanoparticles, and the main analytical methods to confirm this successful conjugation.
Collapse
|
18
|
Veroniaina H, Pan X, Wu Z, Qi X. Apoferritin: a potential nanocarrier for cancer imaging and drug delivery. Expert Rev Anticancer Ther 2021; 21:901-913. [PMID: 33844625 DOI: 10.1080/14737140.2021.1910027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Introduction: As a protein-based biomaterial for potential cancer targeting delivery, apoferritin has recently attracted interest.Areas covered: In this review, we discuss the development of this cage-like protein as an endogenous nanocarrier that can hold molecules in its cavity. We present the specific characterizations and formulations of apoferritin nanocarriers, and outline the recent progress of the protein as an appropriate tumor-delivery vehicle in different therapeutic strategies to treat solid tumors. Finally, we propose how the application for cancer drug repurposing delivery within apoferritin could expand cancer treatment in the future.Expert opinion: Being a ubiquitous iron storage protein that exists in many living organisms, apoferritin is promising as a cancer tumor-targeting nanocarrier. By exploiting its versatility, apoferritin could be used for cancer repurposed drug delivery and could reduce the high cost of new drug discovery development and shorten the formulation process.
Collapse
Affiliation(s)
| | - Xiuhua Pan
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhenghong Wu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
19
|
Lysosomal escaped protein nanocarriers for nuclear-targeted siRNA delivery. Anal Bioanal Chem 2021; 413:3493-3499. [PMID: 33770206 DOI: 10.1007/s00216-021-03297-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/27/2021] [Accepted: 03/16/2021] [Indexed: 01/25/2023]
Abstract
In the process of drug carrier design, lysosome degradation in cells is often neglected, which makes a considerable number of drugs not play a role. Here, we have constructed a tumor treatment platform (Apn/siRNA/NLS/HA/Apt) with unique lysosomal escape function and excellent cancer treatment effect. Apoferritin (Apn) has attracted more and more attention because of its high uniformity, modifiability, and controllability. Meanwhile, its endogenous nature can avoid the risk of immune response being eliminated. We used aptamer modified iron deficient protein nanocages (Apn) to tightly encapsulate the combination of siRNA and NLS (siRNA/NLS) with influenza virus hemagglutinin (HA peptide). After Apn/siRNA/NLS/HA/Apt was targeted into cells, the acidic environment of lysosome led to the cleavage of Apn nanocages, and the release of siRNA/NLS and HA peptide. HA peptide can destroy lysosome membrane, make siRNA/NLS escape lysosome, and enter the nucleus under the action of NLS, resulting in efficient gene silencing effect. This kind of cancer treatment strategy based on Apn nanocage shows high biocompatibility and unique lysosome escape property, which significantly improves the drug delivery and treatment efficiency. Lysosomal escape protein nanocarriers for nuclear-targeted siRNA delivery.
Collapse
|
20
|
Zhang K, Li H, Wang W, Cao J, Gan N, Han H. Application of Multiplexed Aptasensors in Food Contaminants Detection. ACS Sens 2020; 5:3721-3738. [PMID: 33284002 DOI: 10.1021/acssensors.0c01740] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The existence of contaminants in food poses a serious threat to human health. In recent years, aptamer sensors (aptasensors) have been developed rapidly for the detection of food contaminants because of their high specificity, design flexibility, and high efficiency. However, the development of high-throughput, highly sensitive, on-site, and cost-effective methods for simultaneous detection of food contaminants is still restricted due to multiple signal overlap or mutual interference and cross-reaction between different analytes with similar molecular structures. To overcome these problems, this Review summarizes some effective strategies from the articles published in recent years about multiplexed aptasensors for the simultaneous detection of food contaminants. This work focuses on the application of multiplexed aptasensors to simultaneously detect antibiotics, pathogens, and mycotoxins in food. These aptasensors mainly contain fluorescent aptasensors, electrochemical aptasensors, surface-enhanced Raman scattering-based aptasensors, microfluidic chip aptasensors, and paper-based multiplexed aptasensors. In addition, this Review also covers the application of nucleic acid cycle amplification and nanomaterial amplification strategies to improve the detection sensitivity. Finally, the limitations and challenges in the design of multiplexed aptasensor are also taken into account.
Collapse
Affiliation(s)
- Kai Zhang
- The State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Science, Huazhong Agricultural University, Wuhan 430070, Hubei, P.R. China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Faculty of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, P.R. China
| | - Hongyang Li
- College of Life Science, Henan Agricultural University, Zhengzhou 450002, Henan, P.R. China
| | - Wenjing Wang
- The State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Science, Huazhong Agricultural University, Wuhan 430070, Hubei, P.R. China
| | - Jinxuan Cao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Faculty of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, P.R. China
| | - Ning Gan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Faculty of Material Science and Chemical Engineering, Ningbo University, Ningbo 315211, P.R. China
| | - Heyou Han
- The State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Science, Huazhong Agricultural University, Wuhan 430070, Hubei, P.R. China
| |
Collapse
|
21
|
Banstola A, Duwa R, Emami F, Jeong JH, Yook S. Enhanced Caspase-Mediated Abrogation of Autophagy by Temozolomide-Loaded and Panitumumab-Conjugated Poly(lactic-co-glycolic acid) Nanoparticles in Epidermal Growth Factor Receptor Overexpressing Glioblastoma Cells. Mol Pharm 2020; 17:4386-4400. [DOI: 10.1021/acs.molpharmaceut.0c00856] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu 42601, South Korea
| | - Ramesh Duwa
- College of Pharmacy, Keimyung University, Daegu 42601, South Korea
| | | | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Gyeongbuk, South Korea
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu 42601, South Korea
| |
Collapse
|
22
|
Muhetaer A, Niyaz M, Ainiwaer J, Liwei Z, Awut E. Amplification and clinicopathological significance of HER-2 in Kazakh esophageal squamous cell carcinoma. Mol Clin Oncol 2020; 13:64. [PMID: 32968485 PMCID: PMC7500049 DOI: 10.3892/mco.2020.2134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 08/25/2020] [Indexed: 11/06/2022] Open
Abstract
Amplification and overexpression of the human epidermal growth factor receptor-2 (HER-2) gene accelerates cell division and proliferation, and promotes tumor growth and metastasis in various malignant tumors. However, there are few reports on its influence and mechanism in esophageal cancer. The aim of the present study was to investigate the gene amplification and clinicopathological significance of HER-2 in Kazakh esophageal squamous cell carcinoma (ESCC). HER-2 gene amplification was detected in 70 esophageal cancer tissues using fluorescence in situ hybridization. The association between the HER-2 gene amplification and the clinicopathological characteristics of patients with esophageal cancer was also analyzed. The amplification rate of the HER-2 gene in patients with esophageal cancer was 54.2% (38/70). The results also revealed a positive association between the amplification rate of the HER-2 gene in esophageal squamous cell carcinomas and the level of tissue differentiation, increasing gradually and significantly among the highly, moderately and poorly differentiated tissues (P<0.05). The amplification rate of the HER-2 gene in patients with lymph node metastasis was higher than those without (P<0.05). There was no significant association between the amplification rate of the HER-2 gene and any of the clinic pathological parameters, such as sex, age, depth of invasion and 3-year survival, among patients (P>0.05). In conclusion, the amplification rate of the HER-2 gene in patients with Kazakh ESCC was high. There was an association with various prognostic factors, including cancer differentiation and lymph node metastasis. HER-2 gene expression levels may be considered as an indicator of poor prognosis in patients with ESCC in the clinical setting, and this may provide a basis of treatment for individualized targeted therapies.
Collapse
Affiliation(s)
- Aishanjiang Muhetaer
- Department of Thoracic Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China.,Department of Thoracic Surgery, Kashi First People's Hospital, Kashi, Xinjiang 844000, P.R. China
| | - Madiniyet Niyaz
- First Affiliated Hospital/Clinical Medicine Research Institute of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Julaiti Ainiwaer
- Department of Thoracic Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Zhang Liwei
- Department of Thoracic Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Edris Awut
- Department of Thoracic Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| |
Collapse
|
23
|
Zhang C, Zhang X, Zhao G. Ferritin Nanocage: A Versatile Nanocarrier Utilized in the Field of Food, Nutrition, and Medicine. NANOMATERIALS 2020; 10:nano10091894. [PMID: 32971961 PMCID: PMC7557750 DOI: 10.3390/nano10091894] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022]
Abstract
Compared with other nanocarriers such as liposomes, mesoporous silica, and cyclodextrin, ferritin as a typical protein nanocage has received considerable attention in the field of food, nutrition, and medicine owing to its inherent cavity size, excellent water solubility, and biocompatibility. Additionally, ferritin nanocage also serves as a versatile bio-template for the synthesis of a variety of nanoparticles. Recently, scientists have explored the ferritin nanocage structure for encapsulation and delivery of guest molecules such as nutrients, bioactive molecules, anticancer drugs, and mineral metal ions by taking advantage of its unique reversible disassembly and reassembly property and biomineralization. In this review, we mainly focus on the preparation and structure of ferritin-based nanocarriers, and regulation of their self-assembly. Moreover, the recent advances of their applications in food nutrient delivery and medical diagnostics are highlighted. Finally, the main challenges and future development in ferritin-directed nanoparticles’ synthesis and multifunctional applications are discussed.
Collapse
|
24
|
Wang D, Chen L, Wang M, Cui M, Huang L, Xia W, Guan X. Delivering Proapoptotic Peptide by HSP Nanocage for Cancer Therapy. MACROMOL CHEM PHYS 2020. [DOI: 10.1002/macp.202000003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Dongmei Wang
- College of Medical TechnologyBeihua University Jilin 132013 P. R. China
| | - Li Chen
- College of Medical TechnologyBeihua University Jilin 132013 P. R. China
| | - Mingyue Wang
- College of Medical TechnologyBeihua University Jilin 132013 P. R. China
| | - Meiying Cui
- College of Medical TechnologyBeihua University Jilin 132013 P. R. China
| | - Lili Huang
- College of Medical TechnologyBeihua University Jilin 132013 P. R. China
| | - Wei Xia
- College of Medical TechnologyBeihua University Jilin 132013 P. R. China
| | - Xingang Guan
- College of Medical TechnologyBeihua University Jilin 132013 P. R. China
| |
Collapse
|
25
|
Yao H, Guo X, Zhou H, Ren J, Li Y, Duan S, Gong X, Du B. Mild Acid-Responsive "Nanoenzyme Capsule" Remodeling of the Tumor Microenvironment to Increase Tumor Penetration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:20214-20227. [PMID: 32248684 DOI: 10.1021/acsami.0c03022] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Dense extracellular matrix (ECM) severely impedes the spread of drugs in solid tumors and induces hypoxia, reducing chemotherapy efficiency. Different proteolytic enzymes, such as collagenase (Col) or bromelain, can directly attach to the surface of nanoparticles and improve their diffusion, but the method of ligation may also impair the enzymatic activity due to conformational changes or blockage of the active site. Herein, a "nanoenzyme capsule" was constructed by combining collagenase nanocapsules (Col-nc) with heavy-chain ferritin (HFn) nanocages encapsulating the chemotherapy drug doxorubicin (DOX) to enhance tumor penetration of the nanoparticles by hydrolyzing collagen from the ECM. Col-nc could protect the activity of the enzyme before reaching the site of action while being degraded under mildly acidic conditions in tumors, and the released proteolytic enzyme could digest collagen. In addition, HFn as a carrier could effectively load DOX and had a self-targeting ability, enabling the nanoparticles to internalize into cancer cells more effectively. From in vivo and in vitro studies, we found that collagen was effectively degraded by Col-nc/HFn(DOX) to increase the accumulation and penetration of nanoparticles in the solid tumor site and could alleviate hypoxia inside the tumor to enhance the antitumor effects of DOX. Therefore, the strategy of increasing nanoparticle penetration in this system is expected to provide a potential approach for the clinical treatment of solid tumors.
Collapse
Affiliation(s)
- Hanchun Yao
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of Drug Research and Safety Evaluation, Zhengzhou 450001, Henan Province, China
| | - Xiaofang Guo
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Huijuan Zhou
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Jinjin Ren
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Ying Li
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Songchao Duan
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaobao Gong
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Du
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of Drug Research and Safety Evaluation, Zhengzhou 450001, Henan Province, China
| |
Collapse
|
26
|
Yang W, Veroniaina H, Qi X, Chen P, Li F, Ke PC. Soft and Condensed Nanoparticles and Nanoformulations for Cancer Drug Delivery and Repurpose. ADVANCED THERAPEUTICS 2020; 3:1900102. [PMID: 34291146 PMCID: PMC8291088 DOI: 10.1002/adtp.201900102] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Indexed: 12/24/2022]
Abstract
Drug repurpose or reposition is recently recognized as a high-performance strategy for developing therapeutic agents for cancer treatment. This approach can significantly reduce the risk of failure, shorten R&D time, and minimize cost and regulatory obstacles. On the other hand, nanotechnology-based delivery systems are extensively investigated in cancer therapy due to their remarkable ability to overcome drug delivery challenges, enhance tumor specific targeting, and reduce toxic side effects. With increasing knowledge accumulated over the past decades, nanoparticle formulation and delivery have opened up a new avenue for repurposing drugs and demonstrated promising results in advanced cancer therapy. In this review, recent developments in nano-delivery and formulation systems based on soft (i.e., DNA nanocages, nanogels, and dendrimers) and condensed (i.e., noble metal nanoparticles and metal-organic frameworks) nanomaterials, as well as their theranostic applications in drug repurpose against cancer are summarized.
Collapse
Affiliation(s)
- Wen Yang
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | | | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| | - Pengyu Chen
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | - Feng Li
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn AL 36849, USA
| | - Pu Chun Ke
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| |
Collapse
|
27
|
Del Solar V, Contel M. Metal-based antibody drug conjugates. Potential and challenges in their application as targeted therapies in cancer. J Inorg Biochem 2019; 199:110780. [PMID: 31434020 PMCID: PMC6745269 DOI: 10.1016/j.jinorgbio.2019.110780] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/08/2019] [Accepted: 07/14/2019] [Indexed: 12/22/2022]
Abstract
Antibody drug conjugates have emerged as a very attractive type of targeted therapy in cancer. They combine the antigen-targeting specificity of monoclonal antibodies (mAbs) with the cytotoxic potency of chemotherapeutics. This review focuses on antibody drug conjugates based on metal-containing cytotoxic payloads. We will also describe antibody drug conjugates (ADCs) in which a metal-based component (mostly metallic nanoparticles) exerts a relevant function in the ADC (for photodynamic or photothermal therapy, as air-plasma-enhancer or chemo-sensitizer, as carrier of other cytotoxic payloads or as an integral part of the linker structure). Challenges and opportunities to increase the translational potential of these ADCs will be discussed.
Collapse
Affiliation(s)
- Virginia Del Solar
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY 11210, USA
| | - María Contel
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY 11210, USA; Biology PhD Program, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; Biochemistry PhD Program, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; Chemistry PhD Program, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, USA.
| |
Collapse
|
28
|
Akbarzadeh Khiavi M, Safary A, Somi MH. Recent advances in targeted therapy of colorectal cancer: impacts of monoclonal antibodies nanoconjugates. ACTA ACUST UNITED AC 2019; 9:123-127. [PMID: 31508327 PMCID: PMC6726747 DOI: 10.15171/bi.2019.16] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Despite rapid advances in diagnostic and treatment approaches, the overall survival rate of cancer has not been improved. Colorectal cancer (CRC) is recognized as the third leading cause of neoplasm-related deaths worldwide, in large part due to its considerable metastasis and drug resistance. For developing new anticancer strategies, rapid progression of multimodal nanomedicines and nanoconjugates has provided promising treatment modalities for effective therapy of cancer. The limitations of cancer chemotherapy might be overcome through the use of such nanosized therapeutics, including nanoconjugates of monoclonal antibodies (mAbs) along with drugs and organic/inorganic nanoparticles. CRC cells express various molecular markers against which mAbs can be designed and used as targeting/therapeutic agents. This editorial highlights the importance of such targeted nanosystems against CRC.
Collapse
Affiliation(s)
- Mostafa Akbarzadeh Khiavi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, 51656-65811, Iran
| | - Azam Safary
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, 51656-65811, Iran.,Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
29
|
Lin CY, Shieh MJ. Near-Infrared Fluorescent Dye-Decorated Nanocages to Form Grenade-like Nanoparticles with Dual Control Release for Photothermal Theranostics and Chemotherapy. Bioconjug Chem 2018; 29:1384-1398. [PMID: 29505243 DOI: 10.1021/acs.bioconjchem.8b00088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Recently, nanoparticles (NPs) have been widely investigated for delivery of anticancer drugs. Here, a dual control drug-release modality was developed that uses naturally occurring protein apoferritin loaded with doxorubicin (DOX) and ADS-780 near-infrared (NIR) fluorescent dye-decorated NPs (ADNIR NPs). ADNIR NPs act as a grenade to detonate the targeted tumor site following laser irradiation (photothermal therapy, PTT) and explode into cluster warheads (apoferritin-loaded DOX nanocages, AF-DOX NCs) that further destroy the tumor cells (chemotherapy). Light was shown to disrupt the grenade-like structure of NPs to release AF-DOX NCs as well as DOX from NCs in low-pH intercellular environments. In vitro and in vivo studies showed that the structure of AF-DOX NCs was disassembled to release DOX, which then killed the cancer cells in organelles with acidic environments. In vivo studies showed that the ADNIR NP-decorated with NIR dye facilitated tracking of the accumulated NPs at the tumor site using an IVIS imaging system. Overall, targeted ADNIR NPs with dual-release mechanisms were developed for use in photothermal theranostic and chemotherapy. This modality has high potential for application in cancer treatment and clinical translation for drug delivery and imaging.
Collapse
Affiliation(s)
- Chun-Yen Lin
- Institute of Biomedical Engineering, College of Medicine and College of Engineering , National Taiwan University , No. 1, Section 1, Jen-Ai Road , Taipei 100 , Taiwan
| | - Ming-Jium Shieh
- Institute of Biomedical Engineering, College of Medicine and College of Engineering , National Taiwan University , No. 1, Section 1, Jen-Ai Road , Taipei 100 , Taiwan.,Department of Oncology , National Taiwan University Hospital and College of Medicine , #7, Chung-Shan South Road , Taipei 100 , Taiwan
| |
Collapse
|