1
|
Vanni S, Caputo TM, Cusano AM, De Vita A, Cusano A, Cocchi C, Mulè C, Principe S, Liverani C, Celetti G, Micco A, Spadazzi C, Miserocchi G, Ibrahim T, Mercatali L, Aliberti A. Engineered anti-HER2 drug delivery nanosystems for the treatment of breast cancer. NANOSCALE 2025; 17:9436-9457. [PMID: 40116284 DOI: 10.1039/d4nr03907f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Breast cancer stands as the primary cancer affecting women and the second most prevalent cause of cancer-related fatalities in developed nations. Consequently, there is a pressing demand for the advancement of therapeutic strategies that can be seamlessly integrated into clinical applications. We investigated the effectiveness of an encapsulation and decoration strategy employing biodegradable and biocompatible carriers together with 3D collagen-based culture models. Envisioning the use of nano delivery systems for localized regional release, we explored the feasibility of a light-controlled drug release, assisted by optical fibers. PLGA nanoparticles loaded or decorated with trastuzumab (TZ) were synthesized via a double emulsion protocol and characterized by dynamic light scattering, surface plasmon resonance, transmission electron microscopy, atomic force microscopy, and Fourier transform infrared spectroscopy. In vitro biological evaluation was then performed on HER2-positive breast cancer cell line BT-474, examining the effect of nanoformulations on cell viability in 2D and 3D collagen scaffold culture models. Cell cycle, apoptosis, cell morphology and distribution and protein expression were analyzed. Finally, a core-offset optical fiber was fabricated and particles release was studied in vitro using light in batch and microfluidic tests. The nanoparticles displayed uniform and spherical shape, maintaining stability in DMEM for up to seven days. The successful immobilization of TZ was verified. In vitro trials with BT-474 cells in 2D and 3D models revealed that poly(lactic-co-glycolic acid) (PLGA) nanoparticles encapsulated with TZ demonstrated similar or superior biological activity compared to free TZ. Notably, PLGA functionalized with TZ both internally and on the surface exhibited the highest effectiveness in terms of cell viability, increase of apoptosis markers, and inducing cell quiescence. This affirms the pivotal role of PLGA nanoparticles in preserving the integrity of TZ and enhancing its targeted delivery. Furthermore, we propose a breakthrough fiber-optic technology for the less invasive local delivery of PLGA-based nanocarriers that can be effectively used in clinical practice. In conclusion our studies lay the foundation for future advancements in alternative therapeutic tools for localized breast cancer treatment. The integration of advanced carriers, optical fibers, and microfluidics opens up new possibilities for innovative and targeted therapeutic approaches.
Collapse
Affiliation(s)
- Silvia Vanni
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | | | - Angela Maria Cusano
- CeRICTscrl Regional Center Information Communication Technology, Benevento, Italy
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Andrea Cusano
- Optoelectronics Group, Department of Engineering, University of Sannio, Benevento, Italy.
- CeRICTscrl Regional Center Information Communication Technology, Benevento, Italy
| | - Claudia Cocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Chiara Mulè
- Optoelectronics Group, Department of Engineering, University of Sannio, Benevento, Italy.
| | - Sofia Principe
- Optoelectronics Group, Department of Engineering, University of Sannio, Benevento, Italy.
| | - Chiara Liverani
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Giorgia Celetti
- Optoelectronics Group, Department of Engineering, University of Sannio, Benevento, Italy.
| | - Alberto Micco
- CeRICTscrl Regional Center Information Communication Technology, Benevento, Italy
| | - Chiara Spadazzi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Laura Mercatali
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Anna Aliberti
- Optoelectronics Group, Department of Engineering, University of Sannio, Benevento, Italy.
| |
Collapse
|
2
|
Golovina E, Eaton C, Cox V, Andel J, Savvulidi Vargova K. Mechanism of Action of circRNA/miRNA Network in DLBCL. Noncoding RNA 2025; 11:22. [PMID: 40126346 PMCID: PMC11932212 DOI: 10.3390/ncrna11020022] [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: 10/26/2024] [Revised: 02/06/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Circular RNAs (circRNAs) make up approximately 10% of the human transcriptome. CircRNAs belong to the broad group of non-coding RNAs and characteristically are formed by backsplicing into a stable circular loop. Their main role is to regulate transcription through the inhibition of miRNAs' expression, termed miRNA sponging. CircRNAs promote tumorigenesis/lymphomagenesis by competitively binding to miRNAs at miRNA binding sites. In diffuse large B-cell lymphoma (DLBCL), several circRNAs have been identified and their expression is related to both progression and response to therapy. DLBCL is the most prevalent and aggressive subtype of B-cell lymphomas and accounts for about 25% to 30% of all non-Hodgkin lymphomas. DLBCL displays great heterogeneity concerning histopathology, biology, and genetics. Patients who have relapsed or have refractory disease after first-line therapy have a very poor prognosis, demonstrating an important unmet need for new treatment options. As more circRNAs are identified in the future, we will better understand their biological roles and potential use in treating cancer, including DLBCL. For example, circAmotl1 promotes nuclear translocation of MYC and upregulation of translational targets of MYC, thus enhancing lymphomagenesis. Another example is circAPC, which is significantly downregulated in DLBCL and correlates with disease aggressiveness and poor prognosis. CircAPC increases expression of the host gene adenomatous polyposis coli (APC), and in doing so inactivates the canonical Wnt/β-catenin signaling and restrains DLBCL growth. MiRNAs belong to the non-coding regulatory molecules that significantly contribute to lymphomagenesis through their target mRNAs. In DLBCL, among the highly expressed miRNAs, are miR-155-5p and miR-21-5p, which regulate NF-ĸB and PI3K/AKT signaling pathways. The aim of this review is to describe the function and mechanism of regulation of circRNAs on miRNAs' expression in DLBCL. This will help us to better understand the regulatory network of circRNA/miRNA/mRNA, and to propose novel therapeutic targets to treat DLBCL.
Collapse
Affiliation(s)
- Elena Golovina
- First Faculty of Medicine, Institute of Pathological Physiology, Charles University, 12108 Prague, Czech Republic; (E.G.); (C.E.)
| | - Cory Eaton
- First Faculty of Medicine, Institute of Pathological Physiology, Charles University, 12108 Prague, Czech Republic; (E.G.); (C.E.)
| | - Virginia Cox
- First Faculty of Medicine, Institute of Pathological Physiology, Charles University, 12108 Prague, Czech Republic; (E.G.); (C.E.)
| | - Jozef Andel
- First Faculty of Medicine, Institute of Pathological Physiology, Charles University, 12108 Prague, Czech Republic; (E.G.); (C.E.)
- Faculty of Science, Molecular Biology and Genetics of Eukaryotes, Charles University, 12800 Prague, Czech Republic
| | - Karina Savvulidi Vargova
- First Faculty of Medicine, Institute of Pathological Physiology, Charles University, 12108 Prague, Czech Republic; (E.G.); (C.E.)
| |
Collapse
|
3
|
Dai Y, Yu X, Zhao Y, Wei J, Lin D, Wang J, Zhang R, Yuan X, Li S, Huang S, Liu Q, Zhang Z. Targeted Modulation of the Meningeal Lymphatic Reverse Pathway for Immunotherapy of Breast Cancer Brain Metastases. ACS NANO 2025; 19:4830-4844. [PMID: 39818794 DOI: 10.1021/acsnano.4c15860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Treatment of tumor brain metastases remains challenging due to the ineffectiveness of drugs in crossing the blood-brain barrier (BBB). Here, we proposed a potential strategy to target and modulate the meningeal lymphatic system for immunotherapy of breast cancer brain metastases (BCBM) through peripheral administration. CT/fluorescence dual-modality imaging demonstrated that the phospholipid nanoprobe (α-PLNPs) through intracisternal magna injection effectively labeled and long-range tracked the meningeal lymphatic pathway from meningeal lymphatic vessels (MLVs) to periphery drainage cervical lymph nodes (CLNs). Interestingly, the reverse pathway from CLNs to MLVs was also successfully labeled with α-PLNPs through cervical subcutaneous injection, facilitating the noninvasive delivery of immunomodulators to the meningeal lymphatics. Given this, we used melittin-carrying α-M-PLNPs to trigger the modulation of the meningeal lymphatic reverse pathway, which effectively prevents BCBM and prolongs the survival of mice through activating the antigen-presenting cells in the CLNs and promoting the migration of CD8+ T cells into the metastatic brain tumors. This study highlights the potential of the meningeal lymphatic reverse pathway for the immunotherapy of BCBM, which holds great promise for central nervous system disease therapy without the need for drug delivery via BBB.
Collapse
Affiliation(s)
- Yanfeng Dai
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, Hainan 570228, China
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572024, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572024, China
| | - Xiang Yu
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, Hainan 570228, China
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572024, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572024, China
| | - Yifan Zhao
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jianshuang Wei
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Dong Lin
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jialu Wang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Ren Zhang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xuenan Yuan
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Sanmu Li
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572024, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572024, China
| | - Songlin Huang
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, Hainan 570228, China
| | - Qian Liu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572024, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572024, China
| | - Zhihong Zhang
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, Hainan 570228, China
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572024, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572024, China
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|
4
|
Cheng W, Qu H, Yang J, Chen H, Pan Y, Duan Z, Xue X. Hierarchically Engineered Self-Adaptive Nanoplatform Guided Intuitive and Precision Interventions for Deep-Seated Glioblastoma. ACS NANO 2025; 19:557-579. [PMID: 39754309 DOI: 10.1021/acsnano.4c11006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Glioblastoma multiforme (GBM), particularly the deep-seated tumor where surgical removal is not feasible, poses great challenges for clinical treatments due to complicated biological barriers and the risk of damaging healthy brain tissue. Here, we hierarchically engineer a self-adaptive nanoplatform (SAN) that overcomes delivery barriers by dynamically adjusting its structure, surface charge, particle size, and targeting moieties to precisely distinguish between tumor and parenchyma cells. We further devise a SAN-guided intuitive and precision intervention (SGIPi) strategy which obviates the need for sophisticated facilities, skilled operations, and real-time magnetic resonance imaging (MRI) guidance required by current MRI-guided laser or ultrasound interventions. In a preclinical intracranial GBM mouse model, SGIPi-based photodynamic therapy effectively impedes GBM progression with high tumor specificity and significantly extends overall survival. Moreover, the SGIPi potentiates chemotherapy while minimizing adverse effects; it eradicates intracranial GBM lesions in 100% cases solely through Temozolomide chemotherapy. This SGIPi strategy holds potential to improve the clinical management of GBM, with the possibility of extending survival rates and even achieving complete remission, and may inspire research focus from expensive and complex hardware development to simpler, delivery-based GBM interventions.
Collapse
Affiliation(s)
- Wei Cheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haijing Qu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiaojiao Yang
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Han Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuqing Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiran Duan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiangdong Xue
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
5
|
Hoover E, Roy Chowdhury C, Ruggiero OM, Day ES. Conjugation of Antibodies and siRNA Duplexes to Polymer Nanoparticles via Maleimide-Thiol Chemistry. ACS OMEGA 2024; 9:47637-47646. [PMID: 39651074 PMCID: PMC11618400 DOI: 10.1021/acsomega.4c07025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/04/2024] [Accepted: 11/08/2024] [Indexed: 12/11/2024]
Abstract
Polymeric nanoparticles (NPs) have shown great promise as highly modifiable platforms that can be applied across many different disease states. They are advantageous because they can encapsulate a range of hydrophobic and hydrophilic cargoes while having customizable surface properties. Depending on the desired biointerfacing capabilities, the surface of polymeric NPs can be modified with moieties, such as antibodies, peptides, nucleic acids, and more. The work presented here is intended to provide mechanistic insight into how different parameters influence the loading of antibodies, small interfering ribonucleic acids (siRNAs), or both on the surface of poly(lactic-co-glycolic acid) (PLGA) NPs via maleimide-thiol chemistry. Some of the conjugation parameters investigated include the buffer concentration, maleimide to protein ratio, and the addition of an excipient such as Tween-20. Through variation in the concentration of FZD7 antibodies added to the reaction mixture, we established tunable conjugation and found the upper limit of their loading density under the conditions tested. We also confirmed antibody conjugation through two different mechanisms: via a thiol-modified antibody or a thiol-modified poly(ethylene glycol) (PEG) linker. Conjugation of thiolated siRNA duplexes targeting β-catenin was also investigated through variations in both Tween-20 concentration and CaCl2 buffer concentration. Finally, the coconjugation of both antibodies and siRNA duplexes was explored. Overall, this work outlines a basis for tunable biomolecule loading on polymer NPs using maleimide-thiol chemistry and reveals the incredible versatility of polymer NP platforms.
Collapse
Affiliation(s)
- Elise
C. Hoover
- Department
of Biomedical Engineering, University of
Delaware, Newark, DE 19713, United States
| | - Chitran Roy Chowdhury
- Department
of Biomedical Engineering, University of
Delaware, Newark, DE 19713, United States
| | - Olivia M. Ruggiero
- Department
of Biomedical Engineering, University of
Delaware, Newark, DE 19713, United States
| | - Emily S. Day
- Department
of Biomedical Engineering, University of
Delaware, Newark, DE 19713, United States
- Department
of Materials Science and Engineering, University
of Delaware, Newark, DE 19713, United
States
- Helen
F. Graham Cancer Center and Research Institute, Newark, DE 19713, United States
| |
Collapse
|
6
|
Qiao JX, Guo DY, Tian H, Wang ZP, Fan QQ, Tian Y, Sun J, Zhang XF, Zou JB, Cheng JX, Luan F, Zhai BT. Research progress of paclitaxel nanodrug delivery system in the treatment of triple-negative breast cancer. Mater Today Bio 2024; 29:101358. [PMID: 39677523 PMCID: PMC11638641 DOI: 10.1016/j.mtbio.2024.101358] [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: 07/16/2024] [Revised: 10/27/2024] [Accepted: 11/21/2024] [Indexed: 12/17/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, characterized by the loss or low expression of estrogen receptor (ER), human epidermal growth factor receptor 2 (HER2) and progesterone receptor (PR). Due to the lack of clear therapeutic targets, paclitaxel (PTX) is often used as a first-line standard chemotherapy drug for the treatment of high-risk and locally advanced TNBC. PTX is a diterpenoid alkaloid extracted and purified from Taxus plants, functioning as an anticancer agent by inducing and promoting tubulin polymerization, inhibiting spindle formation in cancer cells, and preventing mitosis. However, its clinical application is limited by low solubility and high toxicity. Nanodrug delivery system (NDDS) is one of the feasible methods to improve the water solubility of PTX and reduce side effects. In this review, we summarize the latest advancements in PTX-targeted NDDS, as well as its combination with other codelivery therapies for TNBC treatment. NDDS includes passive targeting, active targeting, stimuli-responsive, codelivery, and multimode strategies. These systems have good prospects in improving the bioavailability of PTX, enhancing tumor targeting, reducing toxicity, controlling drug release, and reverse tumor multidrug resistance (MDR). This review provides valuable insights into the clinical development and application of PTX-targeted NDDS in the treatment of TNBC.
Collapse
Affiliation(s)
- Jia-xin Qiao
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Dong-yan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Huan Tian
- Department of Pharmacy, National Old Pharmacist Inheritance Studio, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, China
| | - Zhan-peng Wang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Qiang-qiang Fan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Yuan Tian
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jing Sun
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Xiao-fei Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jun-bo Zou
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jiang-xue Cheng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Fei Luan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Bing-tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| |
Collapse
|
7
|
Li Y, Gao X, Li Y, Yan S, Zhang Y, Zheng X, Gu Q. Endocytosis: the match point of nanoparticle-based cancer therapy. J Mater Chem B 2024; 12:9435-9458. [PMID: 39192831 DOI: 10.1039/d4tb01227e] [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: 08/29/2024]
Abstract
Nanomedicine has inspired a ground-breaking strategy for cancer therapy. By intelligently assembling diverse moieties to form nanoparticles, numerous functionalities such as controlled release, synergistic efficiency, and in situ killing can be achieved. The emerging nanoparticles have been designed with elevated targeting efficiency as targeting cancer cells is the primary requirement for nanoparticles. However, effective targeting does not guarantee therapeutic effects as endocytosis is a prerequisite for nanoparticles to exert effects. The recent decade has witnessed the rapid development of endocytosis-oriented nanoparticles, and this review subtly analyzes, categorizes, and exemplifies these nanoparticles according to their biological internalization patterns, and the correlation between the endocytosis mechanism and the property of nanoparticles is bridged. Based on the interdisciplinary vision, the present challenges and future perspectives of nanoparticle design for successful endocytosis are discussed, highlighting the potential strategies for the future development of endocytosis-oriented nanoparticles, thus facilitating the endocytosis-oriented strategy from bench to bedside. The undeniable fact is that endocytosis-oriented nanoparticles will definitely bring new blood to the next generation of advanced cancer therapies.
Collapse
Affiliation(s)
- Yonglu Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China.
| | - Xin Gao
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China.
| | - Yapeng Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China.
| | - Shihai Yan
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China.
| | - Yiru Zhang
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-food Processing, Fuli Institute of Food Science, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Xiaodong Zheng
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-food Processing, Fuli Institute of Food Science, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Qing Gu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China.
| |
Collapse
|
8
|
Liang H, Yin G, Feng D, Chen H, Liu X, Li J. Research trends on nanomaterials in triple negative breast cancer (TNBC): a bibliometric analysis from 2010 to 2024. Drug Deliv Transl Res 2024:10.1007/s13346-024-01704-9. [PMID: 39242466 DOI: 10.1007/s13346-024-01704-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2024] [Indexed: 09/09/2024]
Abstract
Breast cancer (BC) is an important cause of cancer-related death in the world. As a subtype of BC with the worst prognosis, triple-negative breast cancer (TNBC) is a serious threat to human life and health. In recent years, there has been an increasing amount of research aimed at designing and developing nanomaterials for the diagnosis and treatment of TNBC. The purpose of this study was to comprehensively evaluate the current status and trend of the application of nanomaterials in TNBC through bibliometric analysis. Studies focusing on nanomaterials and cancer were searched from the Web of Science core collection (WOSCC) database, and relevant literature meeting the inclusion criteria was selected for inclusion in the study. VOSviewer and CiteSpace were used to perform bibliometric and visual analysis of the included publications. A total of 2338 studies were included. Annual publications have increased from 2010 to 2024. China, the United States and India were the leading countries in the field, accounting for 66.1%, 11.5% and 7.2% of publications, respectively. The Chinese Academy of Sciences and Li Yaping were the most influential institutions and authors, respectively. Journal of Controlled Release was considered the most productive journal. Cancer Research was considered to be the most co-cited journal. Drug delivery and anti-cancer mechanisms related to nanomaterials were considered to be the most widely studied aspects, and green synthesis and anti-cancer mechanisms were also recent research hotspots. In this study, the characteristics of publications were summarized, and the most influential countries, institutions, authors, journals, hot spots and trends in the application of nanomaterials in cancer were identified. These findings provide valuable insights into the current state and future direction of this dynamic field.
Collapse
Affiliation(s)
- Hongyi Liang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Dandan Feng
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Hanhan Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Jinan, Shandong, 250014, China
| | - Xiaofei Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Jinan, Shandong, 250014, China
| | - Jingwei Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Jinan, Shandong, 250014, China.
| |
Collapse
|
9
|
Hheidari A, Mohammadi J, Ghodousi M, Mahmoodi M, Ebrahimi S, Pishbin E, Rahdar A. Metal-based nanoparticle in cancer treatment: lessons learned and challenges. Front Bioeng Biotechnol 2024; 12:1436297. [PMID: 39055339 PMCID: PMC11269265 DOI: 10.3389/fbioe.2024.1436297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Cancer, being one of the deadliest diseases, poses significant challenges despite the existence of traditional treatment approaches. This has led to a growing demand for innovative pharmaceutical agents that specifically target cancer cells for effective treatment. In recent years, the use of metal nanoparticles (NPs) as a promising alternative to conventional therapies has gained prominence in cancer research. Metal NPs exhibit unique properties that hold tremendous potential for various applications in cancer treatment. Studies have demonstrated that certain metals possess inherent or acquired anticancer capabilities through their surfaces. These properties make metal NPs an attractive focus for therapeutic development. In this review, we will investigate the applicability of several distinct classes of metal NPs for tumor targeting in cancer treatment. These classes may include gold, silver, iron oxide, and other metals with unique properties that can be exploited for therapeutic purposes. Additionally, we will provide a comprehensive summary of the risk factors associated with the therapeutic application of metal NPs. Understanding and addressing these factors will be crucial for successful clinical translation and to mitigate any potential challenges or failures in the translation of metal NP-based therapies. By exploring the therapeutic potential of metal NPs and identifying the associated risk factors, this review aims to contribute to the advancement of cancer treatment strategies. The anticipated outcome of this review is to provide valuable insights and pave the way for the advancement of effective and targeted therapies utilizing metal NPs specifically for cancer patients.
Collapse
Affiliation(s)
- Ali Hheidari
- Department of Mechanical Engineering, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Javad Mohammadi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Maryam Ghodousi
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, United States
| | - Mohammadreza Mahmoodi
- Bio-microfluidics Lab, Department of Electrical Engineering and Information Technology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Sina Ebrahimi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Esmail Pishbin
- Bio-microfluidics Lab, Department of Electrical Engineering and Information Technology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, Iran
| |
Collapse
|
10
|
Choi SW, Nam JW. Optimal design of synthetic circular RNAs. Exp Mol Med 2024; 56:1281-1292. [PMID: 38871815 PMCID: PMC11263348 DOI: 10.1038/s12276-024-01251-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 06/15/2024] Open
Abstract
Circular RNAs are an unusual class of single-stranded RNAs whose ends are covalently linked via back-splicing. Due to their versatility, the need to express circular RNAs in vivo and in vitro has increased. Efforts have been made to efficiently and precisely synthesize circular RNAs. However, a review on the optimization of the processes of circular RNA design, synthesis, and delivery is lacking. Our review highlights the multifaceted aspects considered when producing optimal circular RNAs and summarizes the available options for each step of exogenous circular RNA design and synthesis, including circularization strategies. Additionally, this review describes several potential applications of circular RNAs.
Collapse
Affiliation(s)
- Seo-Won Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Jin-Wu Nam
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea.
- Bio-BigData Center, Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, 04763, Republic of Korea.
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, 04763, Republic of Korea.
- Hanyang Institute of Advanced BioConvergence, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
11
|
Hervella P, Alonso-Alonso ML, Sampedro-Viana A, Rodríguez-Yáñez M, López-Dequidt I, Pumar JM, Ouro A, Romaus-Sanjurjo D, Campos F, Sobrino T, Castillo J, Leira Y, Iglesias-Rey R. Differential blood-based biomarkers of subcortical and deep brain small vessel disease. Ther Adv Neurol Disord 2024; 17:17562864241243274. [PMID: 38827243 PMCID: PMC11143814 DOI: 10.1177/17562864241243274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/13/2024] [Indexed: 06/04/2024] Open
Abstract
Background Cerebral small vessel disease is the most common cause of lacunar strokes (LS). Understanding LS pathogenesis is vital for predicting disease severity, prognosis, and developing therapies. Objectives To research molecular profiles that differentiate LS in deep brain structures from those in subcortical white matter. Design Prospective case-control study involving 120 patients with imaging-confirmed LS and a 120 control group. Methods We examined the relationship between Alzheimer's disease biomarkers [amyloid beta (Aβ1-40, Aβ1-42)], serum inflammatory marker (interleukin-6, IL-6), and endothelial dysfunction markers [soluble tumor necrosis factor-like weak inducer of apoptosis, and pentraxin-3 (sTWEAK, PTX3)] with respect to LS occurring in deep brain structures and subcortical white matter. In addition, we investigated links between LS, leukoaraiosis presence (white matter hyperintensities, WMHs), and functional outcomes at 3 months. Poor outcome was defined as a modified Rankin scale >2 at 3 months. Results Significant differences were observed in levels of IL-6, PTX3, and sTWEAK between patients with deep lacunar infarcts and those with recent small subcortical infarcts (20.8 versus 15.6 pg/mL, p < 0.001; 7221.3 versus 4624.4 pg/mL, p < 0.0001; 2528.5 versus 1660.5 pg/mL, p = 0.001). Patients with poor outcomes at 3 months displayed notably higher concentrations of these biomarkers compared to those with good outcomes. By contrast, Aβ1-40 and Aβ1-42 were significantly lower in patients with deep LS (p < 0.0001). Aβ1-42 levels were significantly higher in patients with LS in subcortical white matter who had poor outcomes. WMH severity only showed a significant association with deep LS and correlated with sTWEAK (p < 0.0001). Conclusion The pathophysiological mechanisms of lacunar infarcts in deep brain structures seem different from those in the subcortical white matter. As a result, specific therapeutic and preventive strategies should be explored.
Collapse
Affiliation(s)
- Pablo Hervella
- Neuroimaging and Biotechnology Laboratory, Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Maria Luz Alonso-Alonso
- Neuroimaging and Biotechnology Laboratory, Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana Sampedro-Viana
- Neuroimaging and Biotechnology Laboratory, Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Rodríguez-Yáñez
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, Santiago de Compostela, Spain
| | - Iria López-Dequidt
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, Santiago de Compostela, Spain
- Hospital Clínico Universitario de Ferrol, Ferrol, Spain
| | - José M. Pumar
- Neuroimaging and Biotechnology Laboratory, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Neuroradiology, Hospital Clínico Universitario, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Alberto Ouro
- NeuroAging Laboratory, Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Romaus-Sanjurjo
- NeuroAging Laboratory, Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Campos
- Translational Stroke Laboratory, Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Tomás Sobrino
- NeuroAging Laboratory, Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory, Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Yago Leira
- Hospital Clínico Universitario, Rúa Travesa da Choupana, s/n 15706 Santiago de Compostela, Spain
- NeuroAging Laboratory, Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
- Periodontology Unit, Faculty of Medicine and Odontology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Hospital Clínico Universitario, Rúa Travesa da Choupana, s/n 15706 Santiago de Compostela, Spain
- Neuroimaging and Biotechnology Laboratory, Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
12
|
Haidar LL, Bilek M, Akhavan B. Surface Bio-engineered Polymeric Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310876. [PMID: 38396265 DOI: 10.1002/smll.202310876] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Surface bio-engineering of polymeric nanoparticles (PNPs) has emerged as a cornerstone in contemporary biomedical research, presenting a transformative avenue that can revolutionize diagnostics, therapies, and drug delivery systems. The approach involves integrating bioactive elements on the surfaces of PNPs, aiming to provide them with functionalities to enable precise, targeted, and favorable interactions with biological components within cellular environments. However, the full potential of surface bio-engineered PNPs in biomedicine is hampered by obstacles, including precise control over surface modifications, stability in biological environments, and lasting targeted interactions with cells or tissues. Concerns like scalability, reproducibility, and long-term safety also impede translation to clinical practice. In this review, these challenges in the context of recent breakthroughs in developing surface-biofunctionalized PNPs for various applications, from biosensing and bioimaging to targeted delivery of therapeutics are discussed. Particular attention is given to bonding mechanisms that underlie the attachment of bioactive moieties to PNP surfaces. The stability and efficacy of surface-bioengineered PNPs are critically reviewed in disease detection, diagnostics, and treatment, both in vitro and in vivo settings. Insights into existing challenges and limitations impeding progress are provided, and a forward-looking discussion on the field's future is presented. The paper concludes with recommendations to accelerate the clinical translation of surface bio-engineered PNPs.
Collapse
Affiliation(s)
- Laura Libnan Haidar
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Marcela Bilek
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Behnam Akhavan
- School of Physics, University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- School of Engineering, University of Newcastle, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute (HMRI), Precision Medicine Program, New Lambton Heights, NSW, 2305, Australia
| |
Collapse
|
13
|
Zhao Q, Feng J, Liu F, Liang Q, Xie M, Dong J, Zou Y, Ye J, Liu G, Cao Y, Guo Z, Qiao H, Zheng L, Zhao K. Rhizoma Drynariae-derived nanovesicles reverse osteoporosis by potentiating osteogenic differentiation of human bone marrow mesenchymal stem cells via targeting ER α signaling. Acta Pharm Sin B 2024; 14:2210-2227. [PMID: 38799625 PMCID: PMC11119514 DOI: 10.1016/j.apsb.2024.02.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/27/2023] [Accepted: 01/08/2024] [Indexed: 05/29/2024] Open
Abstract
Although various anti-osteoporosis drugs are available, the limitations of these therapies, including drug resistance and collateral responses, require the development of novel anti-osteoporosis agents. Rhizoma Drynariae displays a promising anti-osteoporosis effect, while the effective component and mechanism remain unclear. Here, we revealed the therapeutic potential of Rhizoma Drynariae-derived nanovesicles (RDNVs) for postmenopausal osteoporosis and demonstrated that RDNVs potentiated osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) by targeting estrogen receptor-alpha (ERα). RDNVs, a natural product isolated from fresh Rhizoma Drynariae root juice by differential ultracentrifugation, exhibited potent bone tissue-targeting activity and anti-osteoporosis efficacy in an ovariectomized mouse model. RDNVs, effectively internalized by hBMSCs, enhanced proliferation and ERα expression levels of hBMSC, and promoted osteogenic differentiation and bone formation. Mechanistically, via the ERα signaling pathway, RDNVs facilitated mRNA and protein expression of bone morphogenetic protein 2 and runt-related transcription factor 2 in hBMSCs, which are involved in regulating osteogenic differentiation. Further analysis revealed that naringin, existing in RDNVs, was the active component targeting ERα in the osteogenic effect. Taken together, our study identified that naringin in RDNVs displays exciting bone tissue-targeting activity to reverse osteoporosis by promoting hBMSCs proliferation and osteogenic differentiation through estrogen-like effects.
Collapse
Affiliation(s)
- Qing Zhao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Junjie Feng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fubin Liu
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qianxin Liang
- The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai 519000, China
| | - Manlin Xie
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiaming Dong
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yanfang Zou
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiali Ye
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Guilong Liu
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Department of Blood Transfusion, Guangdong Heyou International Hospital, Foshan 528306, China
| | - Yue Cao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhaodi Guo
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Hongzhi Qiao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lei Zheng
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kewei Zhao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
14
|
Zhao RJ, Zhang WY, Fan XX. Circular RNAs: Potential biomarkers and therapeutic targets for autoimmune diseases. Heliyon 2024; 10:e23694. [PMID: 38205329 PMCID: PMC10776946 DOI: 10.1016/j.heliyon.2023.e23694] [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: 07/25/2023] [Revised: 12/06/2023] [Accepted: 12/09/2023] [Indexed: 01/12/2024] Open
Abstract
The outcomes and prognosis of autoimmune diseases depend on early diagnosis and effective treatments. However, symptoms of early autoimmune diseases are often remarkably similar to many inflammatory diseases, leading to difficulty in precise diagnosis. Circular RNAs (circRNAs) belong to a novel class of endogenous RNAs, functioning as microRNA (miRNA) sponges or participating in protein coding. It has been shown in many studies that patients with autoimmune diseases have aberrant circRNA expression in liquid biopsy samples (such as plasma, saliva, and urine). Thus, circRNAs are potential biomarkers for the diagnosis and prognosis of autoimmune diseases. Moreover, overexpression and depletion of target circRNAs can be utilized as possible therapeutic approaches for treating autoimmune diseases. In this review, we summarized recent progress in the roles of circRNAs in the pathogenesis of autoimmune diseases, including rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, and type 1 diabetes. We also discussed their potential as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | | | - Xing-Xing Fan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau(SAR), China
| |
Collapse
|
15
|
Huang C, Esfani Sarafraz P, Enayati P, Mortazavi Mamaghani E, Babakhanzadeh E, Nazari M. Circular RNAs in renal cell carcinoma: from mechanistic to clinical perspective. Cancer Cell Int 2023; 23:288. [PMID: 37993909 PMCID: PMC10664289 DOI: 10.1186/s12935-023-03128-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023] Open
Abstract
CircRNAs, a special type of noncoding RNAs characterized by their stable structure and unique abilities to form backsplicing loops, have recently attracted the interest of scientists. These RNAs are abundant throughout the body and play important roles such as microRNA sponges, templates for transcription, and regulation of protein translation and RNA-binding proteins. Renal cancer development is highly correlated with abnormal circRNA expression in vivo. CircRNAs are currently considered promising targets for novel therapeutic approaches as well as possible biomarkers for prognosis and diagnosis of various malignancies. Despite our growing understanding of circRNA, numerous questions remain unanswered. Here, we address the characteristics of circRNAs and their function, focusing in particular on their impact on drug resistance, metabolic processes, metastasis, cell growth, and programmed cell death in renal cancer. In addition, the application of circRNAs as prognostic and diagnostic biomarkers will be discussed.
Collapse
Affiliation(s)
- Chunjie Huang
- School of Medicine, Nantong University, Nantong, China
| | | | - Parisa Enayati
- Biological Sciences Department, Northern Illinois University, DeKalb, Illinois, USA
| | | | - Emad Babakhanzadeh
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Nazari
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, 64155-65117, Yazd, Iran.
| |
Collapse
|
16
|
Carney CP, Pandey N, Kapur A, Saadi H, Ong HL, Chen C, Winkles JA, Woodworth GF, Kim AJ. Impact of Targeting Moiety Type and Protein Corona Formation on the Uptake of Fn14-Targeted Nanoparticles by Cancer Cells. ACS NANO 2023; 17:19667-19684. [PMID: 37812740 DOI: 10.1021/acsnano.3c02575] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
The TWEAK receptor, Fn14, is a promising candidate for active targeting of cancer nanotherapeutics to many solid tumor types, including metastatic breast and primary brain cancers. Targeting of therapeutic nanoparticles (NPs) has been accomplished using a range of targeting moieties including monoclonal antibodies and related fragments, peptides, and small molecules. Here, we investigated a full-length Fn14-specific monoclonal antibody, ITEM4, or an ITEM4-Fab fragment as a targeting moiety to guide the development of a clinical formulation. We formulated NPs with varying densities of the targeting moieties while maintaining the decreased nonspecific adhesivity with receptor targeting (DART) characteristics. To model the conditions that NPs experience following intravenous infusion, we investigated the impact of serum exposure in relation to the targeting moiety type and surface density. To further evaluate performance at the cancer cell level, we performed experiments to assess differences in cellular uptake and trafficking in several cancer cell lines using confocal microscopy, imaging flow cytometry, and total internal reflection fluorescence microscopy. We observed that Fn14-targeted NPs exhibit enhanced cellular uptake in Fn14-high compared to Fn14-low cancer cells and that in both cell lines uptake levels were greater than observed with control, nontargeted NPs. We found that serum exposure increased Fn14-targeted NP specificity while simultaneously reducing the total NP uptake. Importantly, serum exposure caused a larger reduction in cancer cell uptake over time when the targeting moiety was an antibody fragment (Fab region of the monoclonal antibody) compared with the full-length monoclonal antibody targeting moiety. Lastly, we uncovered that full monoclonal antibody-targeted NPs enter cancer cells via clathrin-mediated endocytosis and traffic through the endolysosomal pathway. Taken together, these results support a pathway for developing a clinical formulation using a full-length Fn14 monoclonal antibody as the targeting moiety for a DART cancer nanotherapeutic agent.
Collapse
Affiliation(s)
- Christine P Carney
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Nikhil Pandey
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Anshika Kapur
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Hassan Saadi
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Hwei Ling Ong
- Secretory Physiology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Chixiang Chen
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Jeffrey A Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, Maryland 20742, United States
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, Maryland 20742, United States
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| |
Collapse
|
17
|
Kannan S, Cheng VWT. Nanoparticle drug delivery to target breast cancer brain metastasis: Current and future trends. Int J Cancer 2023; 153:1118-1129. [PMID: 37096795 DOI: 10.1002/ijc.34542] [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: 01/02/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/26/2023]
Abstract
Breast cancer brain metastasis (BCBM) is rapidly becoming an impediment to continuing survival gains seen in breast cancer patients. Drug delivery across the blood-brain barrier is the main issue hindering systemic therapy against BCBM. This review details recent advances in nanoparticle (NP) drug delivery systems to target BCBM. Their primary benefits are: enhanced circulating and intra-BCBM drug biodistribution, BCBM targeting through NP functionalization, opportunities for gene manipulation and their theragnostic applications. Multiple NPs have been synthesized to deliver therapeutic HER2 blockade, which is particularly important given HER2-positive breast cancer's tendency to form BCBM. Finally, we review the clinical context in which NP-based therapeutics have been investigated in BCBM patients. While a breakthrough in improving patient outcomes remain awaited, these clinical trials represent positive steps in the changing attitude towards BCBM as a treatable illness. Although multiple challenges remain in the clinical translation of BCBM-directed NP therapies, ongoing research in the field offers promising avenues for novel targeting of this devastating disease.
Collapse
Affiliation(s)
- Siddarth Kannan
- School of Medicine, University of Central Lancashire, Preston, UK
| | - Vinton W T Cheng
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| |
Collapse
|
18
|
Kindt N, Kotecki N, Awada A. Preclinical models to understand the biology and to discover new targets in brain metastases. Curr Opin Oncol 2023; 35:436-440. [PMID: 37551950 DOI: 10.1097/cco.0000000000000963] [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: 08/09/2023]
Abstract
PURPOSE OF REVIEW Incidence of brain metastases increases overtime therefore it is important to rapidly progress in the discovery of new strategies of treatment for these patients. In consequence, more and more preclinical models of brain metastases (BM) are established to study new treatments for melanoma, lung, and breast cancer BM. Here, we reviewed the most recent findings of new drugs assessed in BM mouse preclinical models. RECENT FINDINGS BM are a common metastatic site of several types of solid cancers and can be difficult to treat due to the unique environment of the brain and the blood-brain barrier. Currently, several preclinical models of BM have been demonstrated that new molecular targeted therapies, small metabolic inhibitors, immunotherapies or a combination of these drugs with radiotherapy lead to a reduction of BM growth and an improvement of mouse survival. SUMMARY The use of preclinical models of BM is crucial to discover new treatment strategies for patients with BM. In the last years, some new drugs have been highlighted in preclinical models and are now tested in clinical trials including patients with brain metastases.
Collapse
Affiliation(s)
- Nadège Kindt
- Laboratoire d'Oncologie Clinique et Expérimentale, Faculté de Médecine, Université Libre de Bruxelles, Institut Jules Bordet
| | - Nuria Kotecki
- Oncology Medicine Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Bruxelles, Belgium
| | - Ahmad Awada
- Laboratoire d'Oncologie Clinique et Expérimentale, Faculté de Médecine, Université Libre de Bruxelles, Institut Jules Bordet
- Oncology Medicine Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Bruxelles, Belgium
| |
Collapse
|
19
|
Caputo TM, Cusano AM, Principe S, Cicatiello P, Celetti G, Aliberti A, Micco A, Ruvo M, Tagliamonte M, Ragone C, Minopoli M, Carriero MV, Buonaguro L, Cusano A. Sorafenib-Loaded PLGA Carriers for Enhanced Drug Delivery and Cellular Uptake in Liver Cancer Cells. Int J Nanomedicine 2023; 18:4121-4142. [PMID: 37525693 PMCID: PMC10387258 DOI: 10.2147/ijn.s415968] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/29/2023] [Indexed: 08/02/2023] Open
Abstract
Introduction Currently, conventional treatments of hepatocellular carcinoma (HCC) are not selective enough for tumor tissue and lead to multidrug resistance and drug toxicity. Although sorafenib (SOR) is the standard first-line systemic therapy approved for the clinical treatment of HCC, its poor aqueous solubility and rapid clearance result in low absorption efficiency and severely limit its use for local treatment. Methods Herein, we present the synthesis of biodegradable polymeric Poly (D, L-Lactide-co-glycolide) (PLGA) particles loaded with SOR (PS) by emulsion-solvent evaporation process. The particles are carefully characterized focusing on particle size, surface charge, morphology, drug loading content, encapsulation efficiency, in vitro stability, drug release behaviour and tested on HepG2 cells. Additionally, PLGA particles have been coupled on side emitting optical fibers (seOF) integrated in a microfluidic device for light-triggered local release. Results PS have a size of 248 nm, tunable surface charge and a uniform and spherical shape without aggregation. PS shows encapsulation efficiency of 89.7% and the highest drug loading (8.9%) between the SOR-loaded PLGA formulations. Treating HepG2 cells with PS containing SOR at 7.5 µM their viability is dampened to 40%, 30% and 17% after 48, 129 and 168 hours of incubation, respectively. Conclusion The high PS stability, their sustained release profile and the rapid cellular uptake corroborate the enhanced cytotoxicity effect on HepG2. With the prospect of developing biomedical tools to control the spatial and temporal release of drugs, we successfully demonstrated the potentiality of seOF for light-triggered local release of the carriers. Our prototypical system paves the way to new devices integrating microfluidics, optical fibers, and advanced carriers capable to deliver minimally invasive locoregional cancer treatments.
Collapse
Affiliation(s)
- Tania Mariastella Caputo
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
| | - Angela Maria Cusano
- CeRICTscrl Regional Center Information Communication Technology, Palazzo Ex Poste, Benevento, Italy
| | - Sofia Principe
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
| | - Paola Cicatiello
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
| | - Giorgia Celetti
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
| | - Anna Aliberti
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
| | - Alberto Micco
- CeRICTscrl Regional Center Information Communication Technology, Palazzo Ex Poste, Benevento, Italy
| | - Menotti Ruvo
- Institute of Biostructure and Bioimaging, National Research Council, Naples, Italy
| | - Maria Tagliamonte
- Innovative Immunological Models Unit, Istituto Nazionale Tumori - IRCCS - “Fond G. Pascale”, Naples, Italy
| | - Concetta Ragone
- Innovative Immunological Models Unit, Istituto Nazionale Tumori - IRCCS - “Fond G. Pascale”, Naples, Italy
| | - Michele Minopoli
- Neoplastic Progression Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Naples, Italy
| | - Maria Vincenza Carriero
- Neoplastic Progression Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Naples, Italy
| | - Luigi Buonaguro
- Innovative Immunological Models Unit, Istituto Nazionale Tumori - IRCCS - “Fond G. Pascale”, Naples, Italy
| | - Andrea Cusano
- Optoelectronics Group, Department of Engineering, University of Sannio, Palazzo Dell’ Aquila Bosco Lucarelli, Benevento, Italy
- CeRICTscrl Regional Center Information Communication Technology, Palazzo Ex Poste, Benevento, Italy
| |
Collapse
|
20
|
Azizi M, Jahanban-Esfahlan R, Samadian H, Hamidi M, Seidi K, Dolatshahi-Pirouz A, Yazdi AA, Shavandi A, Laurent S, Be Omide Hagh M, Kasaiyan N, Santos HA, Shahbazi MA. Multifunctional nanostructures: Intelligent design to overcome biological barriers. Mater Today Bio 2023; 20:100672. [PMID: 37273793 PMCID: PMC10232915 DOI: 10.1016/j.mtbio.2023.100672] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/24/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023] Open
Abstract
Over the past three decades, nanoscience has offered a unique solution for reducing the systemic toxicity of chemotherapy drugs and for increasing drug therapeutic efficiency. However, the poor accumulation and pharmacokinetics of nanoparticles are some of the key reasons for their slow translation into the clinic. The is intimately linked to the non-biological nature of nanoparticles and the aberrant features of solid cancer, which together significantly compromise nanoparticle delivery. New findings on the unique properties of tumors and their interactions with nanoparticles and the human body suggest that, contrary to what was long-believed, tumor features may be more mirage than miracle, as the enhanced permeability and retention based efficacy is estimated to be as low as 1%. In this review, we highlight the current barriers and available solutions to pave the way for approved nanoformulations. Furthermore, we aim to discuss the main solutions to solve inefficient drug delivery with the use of nanobioengineering of nanocarriers and the tumor environment. Finally, we will discuss the suggested strategies to overcome two or more biological barriers with one nanocarrier. The variety of design formats, applications and implications of each of these methods will also be evaluated.
Collapse
Affiliation(s)
- Mehdi Azizi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Dental Implants Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Samadian
- Dental Implants Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masoud Hamidi
- Université Libre de Bruxelles (ULB), École Polytechnique de Bruxelles-BioMatter Unit, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Khaled Seidi
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amirhossein Ahmadieh Yazdi
- Department of Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amin Shavandi
- Université Libre de Bruxelles (ULB), École Polytechnique de Bruxelles-BioMatter Unit, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Sophie Laurent
- General, Organic and Biomedical Chemistry Unit, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons – UMONS, Mons, Belgium
| | - Mahsa Be Omide Hagh
- Immunology Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahid Kasaiyan
- Department of Nephrology and Hypertension, University Medical Center Utrecht, 3508 GA, Utrecht, Netherlands
| | - Hélder A. Santos
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
| |
Collapse
|
21
|
Pandey P, Khan F, Choi M, Singh SK, Kang HN, Park MN, Ko SG, Sahu SK, Mazumder R, Kim B. Review deciphering potent therapeutic approaches targeting Notch signaling pathway in breast cancer. Biomed Pharmacother 2023; 164:114938. [PMID: 37267635 DOI: 10.1016/j.biopha.2023.114938] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023] Open
Abstract
In the current period of drug development, natural products have provided an unrivaled supply of anticancer medications. By modifying the cancer microenvironment and various signaling pathways, natural products and their derivatives and analogs play a significant role in cancer treatment. These substances are effective against several signaling pathways, particularly the cell death pathways (apoptosis and autophagy) and embryonic developmental pathways (Notch, Wnt, and Hedgehog pathways). Natural products have a long history, but more research is needed to understand their current function in the research and development of cancer treatments and the potential for natural products to serve as a significant source of therapeutic agents in the future. Several target-specific anticancer medications failed to treat cancer, necessitating research into natural compounds with multiple target properties. To help develop a better treatment plan for managing breast cancer, this review has outlined the anticancerous potential of several therapeutic approaches targeting the notch signaling system in breast tumors.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering & Technology, Greater Noida 201306, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering & Technology, Greater Noida 201306, India.
| | - Min Choi
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, the Republic of Korea
| | - Sujeet Kumar Singh
- Department of Biotechnology, Noida Institute of Engineering & Technology, Greater Noida 201306, India
| | - Han Na Kang
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, the Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, the Republic of Korea
| | - Seong-Gyu Ko
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, the Republic of Korea
| | - Sanjeev Kumar Sahu
- School of pharmaceutical sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rupa Mazumder
- Noida Institute of Engineering & Technology (Pharmacy Institute), Greater Noida 201306, India
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, the Republic of Korea.
| |
Collapse
|
22
|
Xie J, Zhao X, Zhang P, Zhang Y, Cheng R, Zhong Z, Deng C. Codelivery of BCL2 and MCL1 Inhibitors Enabled by Phenylboronic Acid-Functionalized Polypeptide Nanovehicles for Synergetic and Potent Therapy of Acute Myeloid Leukemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204866. [PMID: 36683178 PMCID: PMC10015845 DOI: 10.1002/advs.202204866] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Acute myeloid leukemia (AML) is the most refractory hematologic malignancy characterized by acute onset, rapid progression, and high recurrence rate. Here, codelivery of BCL2 (ABT199) and MCL1 (TW37) inhibitors using phenylboronic acid-functionalized polypeptide nanovehicles to achieve synergetic and potent treatment of AML is adopted. Leveraging the dynamic boronic ester bonds, BN coordination, and π-π stacking, the nanovehicles reveal remarkably efficient and robust drug coencapsulation. ABT199 can induce a series of pro-apoptotic reactions by promoting the dissociation of the pro-apoptotic protein Bim from BCL2, while the released Bim is often captured by MCL1 protein overexpressed in AML. TW37 has a strong inhibitory ability to MCL1, thereby can restrain the depletion of Bim protein. Dual inhibitor-loaded nanoparticles (NPAT) reveal excellent stability, acid/enzyme/H2 O2 -triggered drug release, and significant cytotoxicity toward MOLM-13-Luc and MV-411 AML cells with low half maximal inhibitory concentrations of 1.15 and 7.45 ng mL-1 , respectively. In mice bearing MOLM-13-Luc or MV-411 AML cancer, NPAT reveal significant inhibition of tumor cell infiltration in bone marrow and main organs, potent suppression of tumor growth, and remarkably elevated mouse survival. With facile construction, varying drug combination, superior safety, synergetic efficacy, the phenylboronic acid-functionalized smart nanodrugs hold remarkable potential for AML treatment.
Collapse
Affiliation(s)
- Jiguo Xie
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Xiaofei Zhao
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Peng Zhang
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Yueyue Zhang
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Ru Cheng
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Chao Deng
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| |
Collapse
|
23
|
Yu J, Xie X, Wang L, Liu W, Xu H, Lu X, Li X, Ren J, Li W. Smart Chondroitin Sulfate Micelles for Effective Targeted Delivery of Doxorubicin Against Breast Cancer Metastasis. Int J Nanomedicine 2023; 18:663-677. [PMID: 36798532 PMCID: PMC9926996 DOI: 10.2147/ijn.s398802] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/27/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Metastasis is a major challenge in breast cancer therapy. The successful chemotherapy of breast cancer largely depends on the ability to block the metastatic process. Herein, we designed a dual-targeting and stimuli-responsive drug delivery system for targeted drug delivery against breast cancer metastasis. Methods AS1411 aptamer-modified chondroitin sulfate A-ss-deoxycholic acid (ACSSD) was synthesized, and the unmodified CSSD was used as the control. Chemotherapeutic drug doxorubicin (DOX)-containing ACSSD (D-ACSSD) micelles were prepared by a dialysis method. The ACSSD conjugate was confirmed by Fourier transform infrared spectroscopy (FTIR), nuclear magnetic resonance (NMR), dynamic light scattering (DLS), and transmission electron microscopy (TEM). In vitro cellular uptake and cytotoxicity of D-ACSSD micelles were studied by confocal laser scanning microscopy (CLSM) and MTT assay in breast tumor cells. The inhibition capability of D-ACSSD micelles in cell migration and invasion was carried out in 4T1 cells. In vivo antitumor activity of DOX-containing micelles was investigated in metastatic 4T1-bearing Balb/c mice. Results D-ACSSD and DOX-loaded CSSD (D-CSSD) micelles exhibited high drug encapsulation content and reduction-responsive characteristics. D-ACSSD micelles were spherical in shape. Compared with D-CSSD, D-ACSSD showed higher cellular uptake and more potent killing activity in 4T1 and MDA-MB-231 cells. Additionally, D-ACSSD exhibited stronger inhibitory effects on the invasion and migration of highly metastatic 4T1 cells than unmodified D-CSSD. Among the DOX-containing formulations, D-ACSSD micelles presented the most effective inhibition of tumor growth and lung metastasis in orthotopic 4T1-bearing mice in vivo. It also revealed that ACSSD micelles did not exhibit obvious systemic toxicity. Conclusion The smart D-ACSSD micelles could be a promising delivery system for the therapy of metastatic breast cancer.
Collapse
Affiliation(s)
- Jingmou Yu
- Huzhou Key Laboratory of Medical and Environmental Applications Technologies, School of Life Sciences, Huzhou University, Huzhou, 313000, People’s Republic of China,Jiangxi Provincial Key Laboratory of System Biomedicine, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Xin Xie
- Jiangxi Provincial Key Laboratory of System Biomedicine, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Liangliang Wang
- Affiliated Hospital of Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Wenbo Liu
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Huifeng Xu
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Xiangmei Lu
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Xiaofan Li
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Jin Ren
- Jiangxi Provincial Key Laboratory of System Biomedicine, Jiujiang University, Jiujiang, 332000, People’s Republic of China,School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, 332000, People’s Republic of China,Correspondence: Jin Ren; Weidong Li, Email ;
| | - Weidong Li
- Jiangxi Provincial Key Laboratory of System Biomedicine, Jiujiang University, Jiujiang, 332000, People’s Republic of China,Jiujiang NO.1 People’s Hospital & Water of Life Hospital, Jiujiang, 332000, People’s Republic of China
| |
Collapse
|
24
|
He Z, Xiang W, Fan Q, Wang L, Chao J. A DNA origami nanostructure embedded with NQO1-activated prodrugs for precision drug delivery. Chem Commun (Camb) 2023; 59:912-915. [PMID: 36594872 DOI: 10.1039/d2cc06367k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A rectangle DNA origami nanostructure equipped with doxorubicin-derived prodrugs targeting a tumor cell-specific enzyme (NQO1) is constructed. Combining the high prodrug payload of DNA origami and NQO1-activated chemotherapy, this nanosystem presents therapeutic selectivity for NQO1-overexpressing MCF-7 cells over healthy L02 cells, offering a potent strategy for precision cancer therapy.
Collapse
Affiliation(s)
- Zhimei He
- Smart Health Big Data Analysis and Location Services Engineering Research Center of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts & Telecommunications, Nanjing, 210023, P. R. China.
| | - Wenjie Xiang
- Key Laboratory for Organic Electronics & Information Displays (KLOEID), Jiangsu Key Laboratory for Biosensors Institute of Advanced Materials (IAM) and School of Materials Science and Engineering, Nanjing University of Posts & Telecommunications, Nanjing, 210023, P. R. China
| | - Qin Fan
- Key Laboratory for Organic Electronics & Information Displays (KLOEID), Jiangsu Key Laboratory for Biosensors Institute of Advanced Materials (IAM) and School of Materials Science and Engineering, Nanjing University of Posts & Telecommunications, Nanjing, 210023, P. R. China
| | - Lianhui Wang
- Key Laboratory for Organic Electronics & Information Displays (KLOEID), Jiangsu Key Laboratory for Biosensors Institute of Advanced Materials (IAM) and School of Materials Science and Engineering, Nanjing University of Posts & Telecommunications, Nanjing, 210023, P. R. China
| | - Jie Chao
- Smart Health Big Data Analysis and Location Services Engineering Research Center of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts & Telecommunications, Nanjing, 210023, P. R. China. .,Key Laboratory for Organic Electronics & Information Displays (KLOEID), Jiangsu Key Laboratory for Biosensors Institute of Advanced Materials (IAM) and School of Materials Science and Engineering, Nanjing University of Posts & Telecommunications, Nanjing, 210023, P. R. China
| |
Collapse
|
25
|
Carney CP, Kapur A, Anastasiadis P, Ritzel RM, Chen C, Woodworth GF, Winkles JA, Kim AJ. Fn14-Directed DART Nanoparticles Selectively Target Neoplastic Cells in Preclinical Models of Triple-Negative Breast Cancer Brain Metastasis. Mol Pharm 2023; 20:314-330. [PMID: 36374573 PMCID: PMC11056964 DOI: 10.1021/acs.molpharmaceut.2c00663] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) patients with brain metastasis (BM) face dismal prognosis due to the limited therapeutic efficacy of the currently available treatment options. We previously demonstrated that paclitaxel-loaded PLGA-PEG nanoparticles (NPs) directed to the Fn14 receptor, termed "DARTs", are more efficacious than Abraxane─an FDA-approved paclitaxel nanoformulation─following intravenous delivery in a mouse model of TNBC BM. However, the precise basis for this difference was not investigated. Here, we further examine the utility of the DART drug delivery platform in complementary xenograft and syngeneic TNBC BM models. First, we demonstrated that, in comparison to nontargeted NPs, DART NPs exhibit preferential association with Fn14-positive human and murine TNBC cell lines cultured in vitro. We next identified tumor cells as the predominant source of Fn14 expression in the TNBC BM-immune microenvironment with minimal expression by microglia, infiltrating macrophages, monocytes, or lymphocytes. We then show that despite similar accumulation in brains harboring TNBC tumors, Fn14-targeted DARTs exhibit significant and specific association with Fn14-positive TNBC cells compared to nontargeted NPs or Abraxane. Together, these results indicate that Fn14 expression primarily by tumor cells in TNBC BMs enables selective DART NP delivery to these cells, likely driving the significantly improved therapeutic efficacy observed in our prior work.
Collapse
Affiliation(s)
- Christine P Carney
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Anshika Kapur
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Pavlos Anastasiadis
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Rodney M Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Chixiang Chen
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, Maryland 20742, United States
| | - Jeffrey A Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, Maryland 20742, United States
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| |
Collapse
|
26
|
Zaitseva O, Hoffmann A, Otto C, Wajant H. Targeting fibroblast growth factor (FGF)-inducible 14 (Fn14) for tumor therapy. Front Pharmacol 2022; 13:935086. [PMID: 36339601 PMCID: PMC9634131 DOI: 10.3389/fphar.2022.935086] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Fibroblast growth factor-inducible 14 (Fn14) is a member of the tumor necrosis factor (TNF) receptor superfamily (TNFRSF) and is activated by its ligand TNF-like weak inducer of apoptosis (TWEAK). The latter occurs as a homotrimeric molecule in a soluble and a membrane-bound form. Soluble TWEAK (sTWEAK) activates the weakly inflammatory alternative NF-κB pathway and sensitizes for TNF-induced cell death while membrane TWEAK (memTWEAK) triggers additionally robust activation of the classical NF-κB pathway and various MAP kinase cascades. Fn14 expression is limited in adult organisms but becomes strongly induced in non-hematopoietic cells by a variety of growth factors, cytokines and physical stressors (e.g., hypoxia, irradiation). Since all these Fn14-inducing factors are frequently also present in the tumor microenvironment, Fn14 is regularly found to be expressed by non-hematopoietic cells of the tumor microenvironment and most solid tumor cells. In general, there are three possibilities how the tumor-Fn14 linkage could be taken into consideration for tumor therapy. First, by exploitation of the cancer associated expression of Fn14 to direct cytotoxic activities (antibody-dependent cell-mediated cytotoxicity (ADCC), cytotoxic payloads, CAR T-cells) to the tumor, second by blockade of potential protumoral activities of the TWEAK/Fn14 system, and third, by stimulation of Fn14 which not only triggers proinflammtory activities but also sensitizes cells for apoptotic and necroptotic cell death. Based on a brief description of the biology of the TWEAK/Fn14 system and Fn14 signaling, we discuss the features of the most relevant Fn14-targeting biologicals and review the preclinical data obtained with these reagents. In particular, we address problems and limitations which became evident in the preclinical studies with Fn14-targeting biologicals and debate possibilities how they could be overcome.
Collapse
Affiliation(s)
- Olena Zaitseva
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Annett Hoffmann
- Department of General, Visceral, Transplantation,Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Otto
- Department of General, Visceral, Transplantation,Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- *Correspondence: Harald Wajant,
| |
Collapse
|
27
|
Chen DL, Ma GX, Yang EL, Yang Y, Wang CH, Sun ZC, Liang HQ, Xu XD, Wei JH. Cadinane-type sesquiterpenoid dimeric diastereomers hibisceusones A-C from infected stems of Hibiscus tiliaceus with cytotoxic activity against triple-negative breast cancer cells. Bioorg Chem 2022; 127:105982. [DOI: 10.1016/j.bioorg.2022.105982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/02/2022]
|
28
|
Güner G, Aßfalg M, Zhao K, Dreyer T, Lahiri S, Lo Y, Slivinschi BI, Imhof A, Jocher G, Strohm L, Behrends C, Langosch D, Bronger H, Nimsky C, Bartsch JW, Riddell SR, Steiner H, Lichtenthaler SF. Proteolytically generated soluble Tweak Receptor Fn14 is a blood biomarker for γ-secretase activity. EMBO Mol Med 2022; 14:e16084. [PMID: 36069059 PMCID: PMC9549706 DOI: 10.15252/emmm.202216084] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/29/2022] [Accepted: 08/15/2022] [Indexed: 11/12/2022] Open
Abstract
Fn14 is a cell surface receptor with key functions in tissue homeostasis and injury but is also linked to chronic diseases. Despite its physiological and medical importance, the regulation of Fn14 signaling and turnover is only partly understood. Here, we demonstrate that Fn14 is cleaved within its transmembrane domain by the protease γ‐secretase, resulting in secretion of the soluble Fn14 ectodomain (sFn14). Inhibition of γ‐secretase in tumor cells reduced sFn14 secretion, increased full‐length Fn14 at the cell surface, and enhanced TWEAK ligand‐stimulated Fn14 signaling through the NFκB pathway, which led to enhanced release of the cytokine tumor necrosis factor. γ‐Secretase‐dependent sFn14 release was also detected ex vivo in primary tumor cells from glioblastoma patients, in mouse and human plasma and was strongly reduced in blood from human cancer patients dosed with a γ‐secretase inhibitor prior to chimeric antigen receptor (CAR)‐T‐cell treatment. Taken together, our study demonstrates a novel function for γ‐secretase in attenuating TWEAK/Fn14 signaling and suggests the use of sFn14 as an easily measurable pharmacodynamic biomarker to monitor γ‐secretase activity in vivo.
Collapse
Affiliation(s)
- Gökhan Güner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marlene Aßfalg
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kai Zhao
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Tobias Dreyer
- Department of Gynecology and Obstetrics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Shibojyoti Lahiri
- Protein Analysis Unit, Faculty of Medicine, Biomedical Center, LMU, Martinsried, Germany
| | - Yun Lo
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bianca Ionela Slivinschi
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Axel Imhof
- Protein Analysis Unit, Faculty of Medicine, Biomedical Center, LMU, Martinsried, Germany
| | - Georg Jocher
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Laura Strohm
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, LMU, Munich, Germany
| | - Christian Behrends
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, LMU, Munich, Germany
| | | | - Holger Bronger
- Department of Gynecology and Obstetrics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Jörg W Bartsch
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Stanley R Riddell
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Harald Steiner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Division of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), LMU, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
29
|
Wu H, Zhou H, Zhang W, Jin P, Shi Q, Miao Z, Wang H, Zha Z. Three birds with one stone: co-encapsulation of diclofenac and DL-menthol for realizing enhanced energy deposition, glycolysis inhibition and anti-inflammation in HIFU surgery. J Nanobiotechnology 2022; 20:215. [PMID: 35524259 PMCID: PMC9074192 DOI: 10.1186/s12951-022-01437-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 04/25/2022] [Indexed: 01/12/2023] Open
Abstract
Despite attracting increasing attention in clinic, non-invasive high-intensity focused ultrasound (HIFU) surgery still commonly suffers from tumor recurrence and even matastasis due to the generation of thermo-resistance in non-apoptotic tumor cells and adverse therapy-induced inflammation with enhanced secretion of growth factors in irradiated region. In this work, inspired by the intrinsic property that the expression of thermo-resistant heat shock proteins (HSPs) is highly dependent with adenosine triphosphate (ATP), dual-functionalized diclofenac (DC) with anti-inflammation and glycolysis-inhibition abilities was successfully co-encapsulated with phase-change dl-menthol (DLM) in poly(lactic-co-glycolic acid) nanoparticles (DC/DLM@PLGA NPs) to realize improved HIFU surgery without causing adverse inflammation. Both in vitro and in vivo studies demonstrated the great potential of DC/DLM@PLGA NPs for serving as an efficient synergistic agent for HIFU surgery, which can not only amplify HIFU ablation efficacy through DLM vaporization-induced energy deposition but also simultaneously sensitize tumor cells to hyperthermia by glycolysis inhibition as well as diminished inflammation. Thus, our study provides an efficient strategy for simultaneously improving the curative efficiency and diminishing the harmful inflammatory responses of clinical HIFU surgery.
Collapse
Affiliation(s)
- Haitao Wu
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Hu Zhou
- Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, Guangdong, China
| | - Wenjie Zhang
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Ping Jin
- Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, Guangdong, China.
| | - Qianqian Shi
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Zhaohua Miao
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Hua Wang
- Department of Oncology, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Zhengbao Zha
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China.
| |
Collapse
|
30
|
Picheth GF, Ganzella FADO, Filizzola JO, Canquerino YK, Cardoso GC, Collini MB, Colauto LB, Figueroa-Magalhães MC, Cavalieri EA, Klassen G. Ligand-mediated nanomedicines against breast cancer: a review. Nanomedicine (Lond) 2022; 17:645-664. [PMID: 35438008 DOI: 10.2217/nnm-2021-0473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ligand-mediated targeting represents the cutting edge in precision-guided therapy for several diseases. Surface engineering of nanomedicines with ligands exhibiting selective or tailored affinity for overexpressed biomolecules of a specific disease may increase therapeutic efficiency and reduce side effects and recurrence. This review focuses on newly developed approaches and strategies to improve treatment and overcome the mechanisms associated with breast cancer resistance.
Collapse
Affiliation(s)
- Guilherme F Picheth
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil.,School of Medicine, Pontifical Catholic University of Paraná, Curitiba, Paraná, Brazil
| | | | - João Oc Filizzola
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Yan K Canquerino
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Gabriela C Cardoso
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Michelle B Collini
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Leonardo B Colauto
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | | | - Edneia Asr Cavalieri
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Giseli Klassen
- Department of Basic Pathology, Federal University of Paraná, Curitiba, Paraná, Brazil
| |
Collapse
|
31
|
Kunachowicz D, Ściskalska M, Jakubek M, Kizek R, Kepinska M. Structural changes in selected human proteins induced by exposure to quantum dots, their biological relevance and possible biomedical applications. NANOIMPACT 2022; 26:100405. [PMID: 35560289 DOI: 10.1016/j.impact.2022.100405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/05/2022] [Accepted: 04/27/2022] [Indexed: 06/15/2023]
Abstract
Quantum dots (QDs) are semi-conductor luminescent nanocrystals usually of 2-10 nm diameter, attracting the significant attention in biomedical studies since emerged. Due to their unique optical and electronic properties, i.e. wide absorption spectra, narrow tunable emission bands or stable, bright photoluminescence, QDs seem to be ideally suited for multi-colour, simultaneous bioimaging and cellular labeling at the molecular level as new-generation probes. A highly reactive surface of QDs allows for conjugating them to biomolecules, what enables their direct binding to areas of interest inside or outside the cell for biosensing or targeted delivery. Particularly protein-QDs conjugates are current subjects of research, as features of QDs can be combined with protein specific functionalities and therefore used as a complex in variety of biomedical applications. It is known that QDs are able to interact with cells, organelles and macromolecules of the human body after administration. QDs are reported to cause changes at proteins level, including unfolding and three-dimensional structure alterations which might hamper proteins from performing their physiological functions and thereby limit the use of QD-protein conjugates in vivo. Moreover, these changes may trigger unwanted cellular outcomes as the effect of different signaling pathways activation. In this review, characteristics of QDs interactions with certain human proteins are presented and discussed. Besides that, the following manuscript provides an overview on structural changes of specific proteins exposed to QDs and their biological and biomedical relevance.
Collapse
Affiliation(s)
- Dominika Kunachowicz
- Department of Pharmaceutical Biochemistry, Division of Biomedical and Environmental Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| | - Milena Ściskalska
- Department of Pharmaceutical Biochemistry, Division of Biomedical and Environmental Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Rene Kizek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Marta Kepinska
- Department of Pharmaceutical Biochemistry, Division of Biomedical and Environmental Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland.
| |
Collapse
|
32
|
Zhang D, You Y, Xu Y, Cheng Q, Xiao Z, Chen T, Shi C, Luo L. Facile synthesis of near-infrared responsive on-demand oxygen releasing nanoplatform for precise MRI-guided theranostics of hypoxia-induced tumor chemoresistance and metastasis in triple negative breast cancer. J Nanobiotechnology 2022; 20:104. [PMID: 35246149 PMCID: PMC8896283 DOI: 10.1186/s12951-022-01294-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/03/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Hypoxia is an important factor that contributes to chemoresistance and metastasis in triple negative breast cancer (TNBC), and alleviating hypoxia microenvironment can enhance the anti-tumor efficacy and also inhibit tumor invasion. METHODS A near-infrared (NIR) responsive on-demand oxygen releasing nanoplatform (O2-PPSiI) was successfully synthesized by a two-stage self-assembly process to overcome the hypoxia-induced tumor chemoresistance and metastasis. We embedded drug-loaded poly (lactic-co-glycolic acid) cores into an ultrathin silica shell attached with paramagnetic Gd-DTPA to develop a Magnetic Resonance Imaging (MRI)-guided NIR-responsive on-demand drug releasing nanosystem, where indocyanine green was used as a photothermal converter to trigger the oxygen and drug release under NIR irradiation. RESULTS The near-infrared responsive on-demand oxygen releasing nanoplatform O2-PPSiI was chemically synthesized in this study by a two-stage self-assembly process, which could deliver oxygen and release it under NIR irradiation to relieve hypoxia, improving the therapeutic effect of chemotherapy and suppressed tumor metastasis. This smart design achieves the following advantages: (i) the O2 in this nanosystem can be precisely released by an NIR-responsive silica shell rupture; (ii) the dynamic biodistribution process of O2-PPSiI was monitored in real-time and quantitatively analyzed via sensitive MR imaging of the tumor; (iii) O2-PPSiI could alleviate tumor hypoxia by releasing O2 within the tumor upon NIR laser excitation; (iv) The migration and invasion abilities of the TNBC tumor were weakened by inhibiting the process of EMT as a result of the synergistic therapy of NIR-triggered O2-PPSiI. CONCLUSIONS Our work proposes a smart tactic guided by MRI and presents a valid approach for the reasonable design of NIR-responsive on-demand drug-releasing nanomedicine systems for precise theranostics in TNBC.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
- The Shunde Affiliated Hospital, Jinan University, Foshan, 528300, China
| | - Yuanyuan You
- Department of Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
- Zhuhai Precision Medical Center, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, Guangdong, People's Republic of China
| | - Yuan Xu
- Department of Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Qingqing Cheng
- Department of Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Zeyu Xiao
- Department of Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Tianfeng Chen
- Department of Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China.
- Zhuhai Precision Medical Center, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, Guangdong, People's Republic of China.
| | - Changzheng Shi
- Department of Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China.
| | - Liangping Luo
- Department of Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
33
|
|
34
|
Chen Q, Li Q, Liang Y, Zu M, Chen N, Canup BS, Luo L, Wang C, Zeng L, Xiao B. Natural exosome-like nanovesicles from edible tea flowers suppress metastatic breast cancer via ROS generation and microbiota modulation. Acta Pharm Sin B 2022; 12:907-923. [PMID: 35256954 PMCID: PMC8897038 DOI: 10.1016/j.apsb.2021.08.016] [Citation(s) in RCA: 161] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/22/2021] [Accepted: 07/05/2021] [Indexed: 12/17/2022] Open
Abstract
Although several artificial nanotherapeutics have been approved for practical treatment of metastatic breast cancer, their inefficient therapeutic outcomes, serious adverse effects, and high cost of mass production remain crucial challenges. Herein, we developed an alternative strategy to specifically trigger apoptosis of breast tumors and inhibit their lung metastasis by using natural nanovehicles from tea flowers (TFENs). These nanovehicles had desirable particle sizes (131 nm), exosome-like morphology, and negative zeta potentials. Furthermore, TFENs were found to contain large amounts of polyphenols, flavonoids, functional proteins, and lipids. Cell experiments revealed that TFENs showed strong cytotoxicities against cancer cells due to the stimulation of reactive oxygen species (ROS) amplification. The increased intracellular ROS amounts could not only trigger mitochondrial damage, but also arrest cell cycle, resulting in the in vitro anti-proliferation, anti-migration, and anti-invasion activities against breast cancer cells. Further mice investigations demonstrated that TFENs after intravenous (i.v.) injection or oral administration could accumulate in breast tumors and lung metastatic sites, inhibit the growth and metastasis of breast cancer, and modulate gut microbiota. This study brings new insights to the green production of natural exosome-like nanoplatform for the inhibition of breast cancer and its lung metastasis via i.v. and oral routes.
Collapse
Key Words
- AF633, Alexa Fluor 633-labeled phalloidin
- ALP, alkaline phosphatase
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- BUN, urea nitrogen
- Breast cancer
- CDK, CYCLIN-dependent kinase
- CRE, creatinine
- DAF-FM DA, 4-amino-5-methylamino-2′,7′-difluorofluorescein diacetate
- DAPI, 4′,6-diamidino-2-phenylindole
- DCFH-DA, dichloro-dihydro-fluorescein diacetate
- DGDG, digalactosyl diacylglycerols
- DHE, dihydroethidium
- DLS, dynamic light scattering
- DiO, 3,3′-dioctadecyloxacarbocyanine perchlorate
- DiR, 1,1′-dioctadecyl-3,3,3′′,3′-tetramethylindotricarbocyanine iodide
- EC, epicatechin
- ECG, epicatechin gallate
- EGCG, epigallocatechin gallate
- Exosome-like nanoparticle
- FBS, fetal bovine serum
- GIT, gastrointestinal tract
- H&E, Hematoxylin & Eosin
- HPLC, high-performance liquid chromatography
- Intravenous injection
- LC‒MS, liquid chromatography‒mass spectrometry
- MFI, mean fluorescence intensity
- MGDG, monogalactosyl diacylglycerols
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- Metastasis
- Microbiota modulation
- NO, nitrogen monoxide
- NPs, nanoparticles
- OUT, operational taxonomic unit
- Oral administration
- PA, phosphatidic acids
- PBS, phosphate-buffered saline
- PC, phosphatidylcholines
- PDI, polydispersity index
- PE, phosphatidylethanolamines
- PG, phosphatidylglycerol
- PI, phosphatidylinositol
- PLT, platelets
- PMe, phosphatidylmethanol
- PS, phosphatidylserine
- RBC, red blood cell
- RNS, reactive nitrogen species
- ROS generation
- ROS, reactive oxygen species
- SA, superoxide anion
- SQDG, sulphoquinovosyl diylyceride
- TEM, transmission electron microscopy
- TFENs, exosome-like NPs from tea flowers
- TG, triglyceride
- TUNEL, TdT-mediated dUTP Nick-end labeling
- Tea flower
- WBC, white blood cell
Collapse
|
35
|
St-Denis-Bissonnette F, Khoury R, Mediratta K, El-Sahli S, Wang L, Lavoie JR. Applications of Extracellular Vesicles in Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:451. [PMID: 35053616 PMCID: PMC8773485 DOI: 10.3390/cancers14020451] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and refractory subtype of breast cancer, often occurring in younger patients with poor clinical prognosis. Given the current lack of specific targets for effective intervention, the development of better treatment strategies remains an unmet medical need. Over the last decade, the field of extracellular vesicles (EVs) has grown tremendously, offering immense potential for clinical diagnosis/prognosis and therapeutic applications. While TNBC-EVs have been shown to play an important role in tumorigenesis, chemoresistance and metastasis, they could be repurposed as potential biomarkers for TNBC diagnosis and prognosis. Furthermore, EVs from various cell types can be utilized as nanoscale drug delivery systems (NDDS) for TNBC treatment. Remarkably, EVs generated from specific immune cell subsets have been shown to delay solid tumour growth and reduce tumour burden, suggesting a new immunotherapy approach for TNBC. Intrinsically, EVs can cross the blood-brain barrier (BBB), which holds great potential to treat the brain metastases diagnosed in one third of TNBC patients that remains a substantial clinical challenge. In this review, we present the most recent applications of EVs in TNBC as diagnostic/prognostic biomarkers, nanoscale drug delivery systems and immunotherapeutic agents, as well as discuss the associated challenges and future directions of EVs in cancer immunotherapy.
Collapse
Affiliation(s)
- Frederic St-Denis-Bissonnette
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Rachil Khoury
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Karan Mediratta
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Sara El-Sahli
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Jessie R. Lavoie
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| |
Collapse
|
36
|
Tan Y, Xiong M, Liu Q, Yin Y, Yin X, Liao S, Wang Y, Hu L, Zhang XB. Precisely controlling the cellular internalization of DNA-decorated semiconductor polymer nanoparticles for drug delivery. RSC Adv 2022; 12:31173-31179. [DOI: 10.1039/d2ra05172a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
Precisely controlling the cellular internalization of DNA-decorated semiconductor polymer nanoparticles (SPN-DNA) for drug delivery based on the minimized nonspecific adhesivity to cells.
Collapse
Affiliation(s)
- Ying Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 41008, P. R. China
| | - Mengyi Xiong
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 41008, P. R. China
| | - Qin Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 41008, P. R. China
| | - Yao Yin
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 41008, P. R. China
| | - Xia Yin
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 41008, P. R. China
| | - Shiyi Liao
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 41008, P. R. China
| | - Youjuan Wang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 41008, P. R. China
| | - Ling Hu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 41008, P. R. China
| | - Xiao-Bing Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 41008, P. R. China
| |
Collapse
|
37
|
Carney CP, Pandey N, Kapur A, Woodworth GF, Winkles JA, Kim AJ. Harnessing nanomedicine for enhanced immunotherapy for breast cancer brain metastases. Drug Deliv Transl Res 2021; 11:2344-2370. [PMID: 34716900 PMCID: PMC8568876 DOI: 10.1007/s13346-021-01039-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2021] [Indexed: 12/15/2022]
Abstract
Brain metastases (BMs) are the most common type of brain tumor, and the incidence among breast cancer (BC) patients has been steadily increasing over the past two decades. Indeed, ~ 30% of all patients with metastatic BC will develop BMs, and due to few effective treatments, many will succumb to the disease within a year. Historically, patients with BMs have been largely excluded from clinical trials investigating systemic therapies including immunotherapies (ITs) due to limited brain penetration of systemically administered drugs combined with previous assumptions that BMs are poorly immunogenic. It is now understood that the central nervous system (CNS) is an immunologically distinct site and there is increasing evidence that enhancing immune responses to BCBMs will improve patient outcomes and the efficacy of current treatment regimens. Progress in IT for BCBMs, however, has been slow due to several intrinsic limitations to drug delivery within the brain, substantial safety concerns, and few known targets for BCBM IT. Emerging studies demonstrate that nanomedicine may be a powerful approach to overcome such limitations, and has the potential to greatly improve IT strategies for BMs specifically. This review summarizes the evidence for IT as an effective strategy for BCBM treatment and focuses on the nanotherapeutic strategies currently being explored for BCBMs including targeting the blood-brain/tumor barrier (BBB/BTB), tumor cells, and tumor-supporting immune cells for concentrated drug release within BCBMs, as well as use of nanoparticles (NPs) for delivering immunomodulatory agents, for inducing immunogenic cell death, or for potentiating anti-tumor T cell responses.
Collapse
Affiliation(s)
- Christine P Carney
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Nikhil Pandey
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Anshika Kapur
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jeffrey A Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery and Neurosurgery, University of Maryland School of Medicine, 800 West Baltimore St., Baltimore, MD, 21201, USA.
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA.
- Departments of Neurosurgery, Pharmacology, and Pharmaceutical Sciences, University of Maryland School of Medicine, 655 W Baltimore St., Baltimore, MD, 21201, USA.
| |
Collapse
|
38
|
MacCuaig WM, Fouts BL, McNally MW, Grizzle WE, Chuong P, Samykutty A, Mukherjee P, Li M, Jasinski J, Behkam B, McNally LR. Active Targeting Significantly Outperforms Nanoparticle Size in Facilitating Tumor-Specific Uptake in Orthotopic Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2021; 13:49614-49630. [PMID: 34653338 PMCID: PMC9783196 DOI: 10.1021/acsami.1c09379] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Nanoparticles are widely studied as theranostic vehicles for cancer; however, clinical translation has been limited due to poor tumor specificity. Features that maximize tumor uptake remain controversial, particularly when using clinically relevant models. We report a systematic study that assesses two major features for the impact on tumor specificity, i.e., active vs passive targeting and nanoparticle size, to evaluate relative influences in vivo. Active targeting via the V7 peptide is superior to passive targeting for uptake by pancreatic tumors, irrespective of nanoparticle size, observed through in vivo imaging. Size has a secondary effect on uptake for actively targeted nanoparticles in which 26 nm nanoparticles outperform larger 45 and 73 nm nanoparticles. Nanoparticle size had no significant effect on uptake for passively targeted nanoparticles. Results highlight the superiority of active targeting over nanoparticle size for tumor uptake. These findings suggest a framework for optimizing similar nonaggregate nanoparticles for theranostic treatment of recalcitrant cancers.
Collapse
Affiliation(s)
- William M. MacCuaig
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, 73104, USA
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Benjamin L. Fouts
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, 73104, USA
| | - Molly W McNally
- Department of Surgery, University of Oklahoma, Oklahoma City, OK, 73104, USA
- Department of Cancer Biology, Wake Forest University, Winston-Salem, NC 27157, USA
| | - William E. Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Phillip Chuong
- Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| | - Abhilash Samykutty
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, 73104, USA
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
- Department of Cancer Biology, Wake Forest University, Winston-Salem, NC 27157, USA
| | | | - Min Li
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, 73104, USA
| | - Jacek Jasinski
- Conn Center Materials Characterization, University of Louisville, Louisville, KY 40202, USA
| | - Bahareh Behkam
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Lacey R. McNally
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, 73104, USA
- Department of Surgery, University of Oklahoma, Oklahoma City, OK, 73104, USA
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
- Department of Cancer Biology, Wake Forest University, Winston-Salem, NC 27157, USA
| |
Collapse
|
39
|
Hyun J, Jun S, Lim H, Cho H, You SH, Ha SJ, Min JJ, Bang D. Engineered Attenuated Salmonella typhimurium Expressing Neoantigen Has Anticancer Effects. ACS Synth Biol 2021; 10:2478-2487. [PMID: 34525796 DOI: 10.1021/acssynbio.1c00097] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Neoantigen vaccines are an immunotherapy strategy for treating cancer. The vaccine degrades quickly, so the strategy must include protection and precise targeting for immune cell stimulation. In this study, we engineered attenuated Salmonella typhimurium, which is highly infiltrative to tumors, to act as a carrier for Neoantigen peptide vaccine. Our system used a constitutive promoter vector, so that a single injection of Salmonella expressing Neoantigen could be used without requiring additional induction injections. In vivo experiments on bacteria-treated mice showed that Neoantigen expressed by the engineered carrier infiltrated tumors and resulted in suppressed tumor growth, higher survival rates and longer survival times, a relative increase of CD4 and CD8 T cells, and cytokine release. These results indicate that engineered Salmonella can be used as a carrier for Neoantigen immunotherapy.
Collapse
Affiliation(s)
- Jungheun Hyun
- Department of Chemistry, Yonsei University, Seoul, 03722, Republic of Korea
| | - Soyeong Jun
- Department of Chemistry, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyeonseob Lim
- Department of Chemistry, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyunjun Cho
- Department of Chemistry, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sung-Hwan You
- Laboratory of In Vivo Molecular Imaging, Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam, 58128, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jung-Joon Min
- Laboratory of In Vivo Molecular Imaging, Institute for Molecular Imaging and Theranostics, Chonnam National University Hwasun Hospital, Jeonnam, 58128, Republic of Korea
| | - Duhee Bang
- Department of Chemistry, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
40
|
Niu W, Guo Y, Xue Y, Wang M, Chen M, Winston DD, Cheng W, Lei B. Biodegradable multifunctional bioactive Eu-Gd-Si-Ca glass nanoplatform for integrative imaging-targeted tumor therapy-recurrence inhibition-tissue repair. NANO TODAY 2021; 38:101137. [DOI: 10.1016/j.nantod.2021.101137] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
|
41
|
He AT, Liu J, Li F, Yang BB. Targeting circular RNAs as a therapeutic approach: current strategies and challenges. Signal Transduct Target Ther 2021; 6:185. [PMID: 34016945 PMCID: PMC8137869 DOI: 10.1038/s41392-021-00569-5] [Citation(s) in RCA: 317] [Impact Index Per Article: 79.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/28/2021] [Accepted: 03/16/2021] [Indexed: 02/04/2023] Open
Abstract
Significant progress has been made in circular RNA (circRNA) research in recent years. Increasing evidence suggests that circRNAs play important roles in many cellular processes, and their dysregulation is implicated in the pathogenesis of various diseases. CircRNAs are highly stable and usually expressed in a tissue- or cell type-specific manner. Therefore, they are currently being explored as potential therapeutic targets. Gain-of-function and loss-of-function approaches are typically performed using circRNA expression plasmids and RNA interference-based strategies, respectively. These strategies have limitations that can be mitigated using nanoparticle and exosome delivery systems. Furthermore, recent developments show that the cre-lox system can be used to knockdown circRNAs in a cell-specific manner. While still in the early stages of development, the CRISPR/Cas13 system has shown promise in knocking down circRNAs with high specificity and efficiency. In this review, we describe circRNA properties and functions and highlight their significance in disease. We summarize strategies that can be used to overexpress or knockdown circRNAs as a therapeutic approach. Lastly, we discuss major challenges and propose future directions for the development of circRNA-based therapeutics.
Collapse
Affiliation(s)
- Alina T. He
- grid.17063.330000 0001 2157 2938Sunnybrook Research Institute, Toronto, ON Canada
| | - Jinglei Liu
- Department of Bioinformatics, ATCGene Inc, Guangzhou, China
| | - Feiya Li
- grid.17063.330000 0001 2157 2938Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| | - Burton B. Yang
- grid.17063.330000 0001 2157 2938Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| |
Collapse
|
42
|
Song S, Xia H, Guo M, Wang S, Zhang S, Ma P, Jin Y. Role of macrophage in nanomedicine-based disease treatment. Drug Deliv 2021; 28:752-766. [PMID: 33860719 PMCID: PMC8079019 DOI: 10.1080/10717544.2021.1909175] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Macrophages are a major component of the immunoresponse. Diversity and plasticity are two of the hallmarks of macrophages, which allow them to act as proinflammatory, anti-inflammatory, and homeostatic agents. Research has found that cancer and many inflammatory or autoimmune disorders are correlated with activation and tissue infiltration of macrophages. Recent developments in macrophage nanomedicine-based disease treatment are proving to be timely owing to the increasing inadequacy of traditional treatment. Here, we review the role of macrophages in nanomedicine-based disease treatment. First, we present a brief background on macrophages and nanomedicine. Then, we delve into applications of macrophages as a target for disease treatment and delivery systems and summarize the applications of macrophage-derived extracellular vesicles. Finally, we provide an outlook on the clinical utility of macrophages in nanomedicine-based disease treatment.
Collapse
Affiliation(s)
- Siwei Song
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Xia
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sufei Wang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujing Zhang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Böhmer VI, Szymanski W, Feringa BL, Elsinga PH. Multivalent Probes in Molecular Imaging: Reality or Future? Trends Mol Med 2021; 27:379-393. [PMID: 33436332 DOI: 10.1016/j.molmed.2020.12.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/17/2020] [Accepted: 12/08/2020] [Indexed: 01/25/2023]
Abstract
The rapidly developing field of molecular medical imaging focuses on specific visualization of (patho)physiological processes through the application of imaging agents (IAs) in multiple clinical modalities. Although our understanding of the principles underlying efficient IAs design has increased tremendously, many IAs still show poor in vivo imaging performance because of low binding affinity and/or specificity. These limitations can be addressed by taking advantage of multivalency, in which multiple copies of a ligand are employed to strengthen the interaction. We critically address specific challenges associated with the application of multivalent compounds in molecular imaging, and we give directions for a stepwise approach to the design of multivalent imaging probes to improve their target binding and pharmacokinetics (PK) for improved diagnostic potential.
Collapse
Affiliation(s)
- Verena I Böhmer
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, The Netherlands; Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747, AF, Groningen, The Netherlands
| | - Wiktor Szymanski
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747, AF, Groningen, The Netherlands; Department of Radiology, Medical Imaging Center, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, The Netherlands
| | - Ben L Feringa
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747, AF, Groningen, The Netherlands
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, The Netherlands.
| |
Collapse
|
44
|
Lv Y, Ma X, Du Y, Feng J. Understanding Patterns of Brain Metastasis in Triple-Negative Breast Cancer and Exploring Potential Therapeutic Targets. Onco Targets Ther 2021; 14:589-607. [PMID: 33519208 PMCID: PMC7837592 DOI: 10.2147/ott.s293685] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly malignant subtype of breast cancer. High invasiveness and heterogeneity, as well as a lack of drug targets, are the main factors leading to poor prognosis. Brain metastasis (BM) is a serious event threatening the life of breast cancer patients, especially those with TNBC. Compared with that for hormone receptor-positive and HER2-positive breast cancers, TNBC-derived BM (TNBCBM) occurs earlier and more frequently, and has a worse prognosis. There is no standard treatment for BM to date, and one is urgently required. In this review, we discuss the current knowledge regarding the developmental patterns of TNBCBM, focusing on the key events in BM formation. Specifically, we consider (i) the nature and function of TNBC cells; (ii) how TNBC cells cross the blood–brain barrier and form a fenestrated, more permeable blood–tumor barrier; (iii) the biological characteristics of TNBCBM; and (iv) the infiltration and colonization of the central nervous system (CNS) by TNBC cells, including the establishment of premetastatic niches, immunosurveillance escape, and metabolic adaptations. We also discuss putative therapeutic targets and precision therapy with the greatest potential to treat TNBCBM, and summarize the relevant completed and ongoing clinical trials. These findings may provide new insights into the prevention and treatment of BM in TNBC patients.
Collapse
Affiliation(s)
- Yan Lv
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing 210009, People's Republic of China
| | - Xiao Ma
- Department of General Surgery, The Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, People's Republic of China
| | - Yuxin Du
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing 210009, People's Republic of China
| | - Jifeng Feng
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing 210009, People's Republic of China
| |
Collapse
|
45
|
Shen J, Cai W, Ma Y, Xu R, Huo Z, Song L, Qiu X, Zhang Y, Li A, Cao W, Zhou S, Tang X. hGC33-Modified and Sorafenib-Loaded Nanoparticles have a Synergistic Anti-Hepatoma Effect by Inhibiting Wnt Signaling Pathway. NANOSCALE RESEARCH LETTERS 2020; 15:220. [PMID: 33242103 PMCID: PMC7691417 DOI: 10.1186/s11671-020-03451-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/12/2020] [Indexed: 05/06/2023]
Abstract
Delivery of tumor-specific inhibitors is a challenge in cancer treatment. Antibody-modified nanoparticles can deliver their loaded drugs to tumor cells that overexpress specific tumor-associated antigens. Here, we constructed sorafenib-loaded polyethylene glycol-b-PLGA polymer nanoparticles modified with antibody hGC33 to glypican-3 (GPC3 +), a membrane protein overexpressed in hepatocellular carcinoma. We found that hGC33-modified NPs (hGC33-SFB-NP) targeted GPC3+ hepatocellular carcinoma (HCC) cells by specifically binding to GPC3 on the surface of HCC cells, inhibited Wnt-induced signal transduction, and inhibited HCC cells in G0/1 by down-regulating cyclin D1 expression, thus attenuating HCC cell migration by inhibiting epithelial-mesenchymal transition. hGC33-SFB-NP inhibited the migration, cycle progression, and proliferation of HCC cells by inhibiting the Ras/Raf/MAPK pathway and the Wnt pathway in tandem with GPC3 molecules, respectively. hGC33-SFB-NP inhibited the growth of liver cancer in vivo and improved the survival rate of tumor-bearing mice. We conclude that hGC33 increases the targeting of SFB-NP to HCC cells. hGC33-SFB-NP synergistically inhibits the progression of HCC by blocking the Wnt pathway and the Ras/Raf/MAPK pathway.
Collapse
Affiliation(s)
- Jing Shen
- Medical School, Anhui University of Science and Technology, Huainan, 232001, China
- Wuhu Research Institute, Anhui University of Science and Technology, Huainan, 232001, China
| | - Wenpeng Cai
- Medical School, Anhui University of Science and Technology, Huainan, 232001, China
- Wuhu Research Institute, Anhui University of Science and Technology, Huainan, 232001, China
| | - Yongfang Ma
- Medical School, Anhui University of Science and Technology, Huainan, 232001, China
- Wuhu Research Institute, Anhui University of Science and Technology, Huainan, 232001, China
| | - Ruyue Xu
- Medical School, Anhui University of Science and Technology, Huainan, 232001, China
- Wuhu Research Institute, Anhui University of Science and Technology, Huainan, 232001, China
| | - Zhen Huo
- Medical School, Anhui University of Science and Technology, Huainan, 232001, China
- Wuhu Research Institute, Anhui University of Science and Technology, Huainan, 232001, China
| | - Li Song
- Medical School, Anhui University of Science and Technology, Huainan, 232001, China
- Wuhu Research Institute, Anhui University of Science and Technology, Huainan, 232001, China
| | - Xinyin Qiu
- Medical School, Anhui University of Science and Technology, Huainan, 232001, China
- Wuhu Research Institute, Anhui University of Science and Technology, Huainan, 232001, China
| | - Yinci Zhang
- Medical School, Anhui University of Science and Technology, Huainan, 232001, China
- Wuhu Research Institute, Anhui University of Science and Technology, Huainan, 232001, China
| | - Amin Li
- Medical School, Anhui University of Science and Technology, Huainan, 232001, China
- Wuhu Research Institute, Anhui University of Science and Technology, Huainan, 232001, China
| | - Weiya Cao
- Medical School, Anhui University of Science and Technology, Huainan, 232001, China
- Wuhu Research Institute, Anhui University of Science and Technology, Huainan, 232001, China
| | - Shuping Zhou
- Medical School, Anhui University of Science and Technology, Huainan, 232001, China
| | - Xiaolong Tang
- Wuhu Research Institute, Anhui University of Science and Technology, Huainan, 232001, China.
| |
Collapse
|
46
|
Mó I, Alves CG, de Melo-Diogo D, Lima-Sousa R, Correia IJ. Assessing the Combinatorial Chemo-Photothermal Therapy Mediated by Sulfobetaine Methacrylate-Functionalized Nanoparticles in 2D and 3D In Vitro Cancer Models. Biotechnol J 2020; 15:e2000219. [PMID: 33063471 DOI: 10.1002/biot.202000219] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Combinatorial cancer therapies mediated by nanomaterials can potentially overcome the limitations of conventional treatments. These therapies are generally investigated using 2D in vitro cancer models, leading to an inaccurate screening. Recently, 3D in vitro spheroids have emerged in the preclinical testing stage of nanomedicines due to their ability to mimic key features of the in vivo solid tumors. Investigate the chemo-photothermal therapy mediated by Doxorubicin and IR780 loaded sulfobetaine methacrylate functionalized nanoparticles, for the first time, using monolayers of cancer cells and spheroids. In the 2D cancer models, the nanomaterials' mediated photothermal therapy, chemotherapy, and chemo-photothermal therapy reduced cancer cells' viability to about 58%, 29%, and 1%, respectively. Interestingly, when the nanomaterials' mediated photothermal therapy is tested on 3D spheroids, no cytotoxic effect is noticed. In contrast, the nanostructures' induced chemotherapy decreased spheroids' viability to 42%. On the other hand, nanomaterials' mediated chemo-photothermal therapy diminished spheroids' viability to 16%, being the most promising therapeutic modality. These results demonstrate the importance of using 3D spheroids during the in vitro screening of single/combinatorial therapies mediated by nanomaterials.
Collapse
Affiliation(s)
- Inês Mó
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, 6200-506, Portugal
| | - Cátia G Alves
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, 6200-506, Portugal
| | - Duarte de Melo-Diogo
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, 6200-506, Portugal
| | - Rita Lima-Sousa
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, 6200-506, Portugal
| | - Ilídio J Correia
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, 6200-506, Portugal.,CIEPQPF - Departamento de Engenharia Química, Rua Sílvio Lima, Universidade de Coimbra, Coimbra, 3030-790, Portugal
| |
Collapse
|
47
|
Wei QY, Xu YM, Lau ATY. Recent Progress of Nanocarrier-Based Therapy for Solid Malignancies. Cancers (Basel) 2020; 12:E2783. [PMID: 32998391 PMCID: PMC7600685 DOI: 10.3390/cancers12102783] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 02/05/2023] Open
Abstract
Conventional chemotherapy is still an important option of cancer treatment, but it has poor cell selectivity, severe side effects, and drug resistance. Utilizing nanoparticles (NPs) to improve the therapeutic effect of chemotherapeutic drugs has been highlighted in recent years. Nanotechnology dramatically changed the face of oncology by high loading capacity, less toxicity, targeted delivery of drugs, increased uptake to target sites, and optimized pharmacokinetic patterns of traditional drugs. At present, research is being envisaged in the field of novel nano-pharmaceutical design, such as liposome, polymer NPs, bio-NPs, and inorganic NPs, so as to make chemotherapy effective and long-lasting. Till now, a number of studies have been conducted using a wide range of nanocarriers for the treatment of solid tumors including lung, breast, pancreas, brain, and liver. To provide a reference for the further application of chemodrug-loaded nanoformulations, this review gives an overview of the recent development of nanocarriers, and the updated status of their use in the treatment of several solid tumors.
Collapse
Affiliation(s)
| | | | - Andy T. Y. Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, Guangdong, China; (Q.-Y.W.); (Y.-M.X.)
| |
Collapse
|
48
|
Lussier F, Staufer O, Platzman I, Spatz JP. Can Bottom-Up Synthetic Biology Generate Advanced Drug-Delivery Systems? Trends Biotechnol 2020; 39:445-459. [PMID: 32912650 DOI: 10.1016/j.tibtech.2020.08.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023]
Abstract
Creating a magic bullet that can selectively kill cancer cells while sparing nearby healthy cells remains one of the most ambitious objectives in pharmacology. Nanomedicine, which relies on the use of nanotechnologies to fight disease, was envisaged to fulfill this coveted goal. Despite substantial progress, the structural complexity of therapeutic vehicles impedes their broad clinical application. Novel modular manufacturing approaches for engineering programmable drug carriers may be able to overcome some fundamental limitations of nanomedicine. We discuss how bottom-up synthetic biology principles, empowered by microfluidics, can palliate current drug carrier assembly limitations, and we demonstrate how such a magic bullet could be engineered from the bottom up to ultimately improve clinical outcomes for patients.
Collapse
Affiliation(s)
- Felix Lussier
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany; Institute for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, 69120 Heidelberg, Germany.
| | - Oskar Staufer
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany; Institute for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, 69120 Heidelberg, Germany; Max Planck-Bristol Centre for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol BS8 1TD, UK; Max Planck School Matter to Life, Jahnstraße 29, 69120 Heidelberg, Germany
| | - Ilia Platzman
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany; Institute for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, 69120 Heidelberg, Germany; Max Planck-Bristol Centre for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol BS8 1TD, UK.
| | - Joachim P Spatz
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany; Institute for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, 69120 Heidelberg, Germany; Max Planck-Bristol Centre for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol BS8 1TD, UK; Max Planck School Matter to Life, Jahnstraße 29, 69120 Heidelberg, Germany.
| |
Collapse
|
49
|
Pinals RL, Chio L, Ledesma F, Landry MP. Engineering at the nano-bio interface: harnessing the protein corona towards nanoparticle design and function. Analyst 2020; 145:5090-5112. [PMID: 32608460 PMCID: PMC7439532 DOI: 10.1039/d0an00633e] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Unpredictable and uncontrollable protein adsorption on nanoparticles remains a considerable challenge to achieving effective application of nanotechnologies within biological environments. Nevertheless, engineered nanoparticles offer unprecedented functionality and control in probing and altering biological systems. In this review, we highlight recent advances in harnessing the "protein corona" formed on nanoparticles as a handle to tune functional properties of the protein-nanoparticle complex. Towards this end, we first review nanoparticle properties that influence protein adsorption and design strategies to facilitate selective corona formation, with the corresponding characterization techniques. We next focus on literature detailing corona-mediated functionalities, including stealth to avoid recognition and sequestration while in circulation, targeting of predetermined in vivo locations, and controlled activation once localized to the intended biological compartment. We conclude with a discussion of biocompatibility outcomes for these protein-nanoparticle complexes applied in vivo. While formation of the nanoparticle-corona complex may impede our control over its use for the projected nanobiotechnology application, it concurrently presents an opportunity to create improved protein-nanoparticle architectures by exploiting natural or guiding selective protein adsorption to the nanoparticle surface.
Collapse
Affiliation(s)
- Rebecca L Pinals
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, USA.
| | | | | | | |
Collapse
|
50
|
Bhargav AG, Mondal SK, Garcia CA, Green JJ, Quiñones‐Hinojosa A. Nanomedicine Revisited: Next Generation Therapies for Brain Cancer. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Adip G. Bhargav
- Mayo Clinic College of Medicine and Science Mayo Clinic 200 First Street SW Rochester MN 55905 USA
- Department of Neurologic Surgery Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
| | - Sujan K. Mondal
- Department of Pathology University of Pittsburgh School of Medicine 200 Lothrop Street Pittsburgh PA 15213 USA
| | - Cesar A. Garcia
- Department of Neurologic Surgery Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
| | - Jordan J. Green
- Departments of Biomedical Engineering, Neurosurgery, Oncology, Ophthalmology, Materials Science and Engineering, and Chemical and Biomolecular Engineering, Translational Tissue Engineering Center, Bloomberg‐Kimmel Institute for Cancer Immunotherapy, Institute for Nanobiotechnology Johns Hopkins University School of Medicine 400 N. Broadway, Smith 5017 Baltimore MD 21231 USA
| | - Alfredo Quiñones‐Hinojosa
- Department of Neurologic Surgery Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
- Departments of Otolaryngology‐Head and Neck Surgery/Audiology Neuroscience, Cancer Biology, and Anatomy Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
| |
Collapse
|