1
|
Huang Y, Chen L, Chen Y, Zhou S, Xie X, Xie J, Yu M, Chen J. High-density lipoprotein-based nanoplatform reprograms tumor microenvironment and enhances chemotherapy against pancreatic ductal adenocarcinoma. Biomaterials 2025; 318:123147. [PMID: 39908877 DOI: 10.1016/j.biomaterials.2025.123147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/25/2024] [Accepted: 01/26/2025] [Indexed: 02/07/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly aggressive, with limited success in traditional therapies due to the fibrotic, immunosuppressive, pro-metastatic tumor microenvironment (TME), which collectively impede the drug accumulation and accelerate the tumor progression. In this work, we developed a PDAC-customized nutrient-mimicking reconstituted high-density lipoprotein (rHDL) capable of efficiently co-encapsulate versatile TME regulating cannabidiol and cytotoxic gemcitabine to simultaneously reprogram TME while suppressing PDAC progression. Specifically, a small-sized, nutrient-like rHDL was constructed to realize deep PDAC parenchyma penetration and efficient intra-tumoral uptake. Next, natural herbal compound cannabidiol was screened and incorporated into rHDL to regulate TME via attenuating fibrosis, reliving immunosuppression and mitigating metastatic tendency. At last, gemcitabine, the PDAC gold standard first-line therapy was co-delivered by the PDAC-customized rHDL to overcome drug resistance and amplify its PDAC suppression. Our findings demonstrate the feasibility of an integrated multi-stage TME regulation strategy for improved PDAC therapy, and might represent a modality in promoting chemotherapy against PDAC.
Collapse
Affiliation(s)
- Yukun Huang
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China; Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liang Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Yu Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Songlei Zhou
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Xiaoying Xie
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Jing Xie
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Minghua Yu
- Fudan University Clinical Research Center for Cell-based Immunotherapy & Department of Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Shanghai, 201399, China
| | - Jun Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
2
|
Zhang W, Chen C, Wang R, Li Y, Zhu Y, Sun J, Luo Y, Luo J, Zhao L, Sun Y, Guo H, Zhang H, Fang B, Hu Y, Liu R, Wang X, Guo J, Liu P, An P, Wang P, Chen J, Ren F. Fe 2+ Triggers Endocytosis of Carrier Proteins in Mammalian Cells. Adv Healthc Mater 2025:e2502090. [PMID: 40420645 DOI: 10.1002/adhm.202502090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2025] [Revised: 05/20/2025] [Indexed: 05/28/2025]
Abstract
Human serum albumin (HSA) plays an indispensable role as a carrier of endogenous and exogenous substances in vivo. The efficiency of endocytosis determines the transport capacity. However, the potential for enhancing the efficiency of HSA endocytosis remains largely unknown. This research represented a substantial advancement, revealing that Fe2+ markedly increased HSA endocytosis. Moreover, Fe2+ facilitated the endocytosis and transport of HSA in vivo. The enhancement in HSA uptake facilitated by Fe2+ is markedly decreased following iron chelation, indicating a specific interaction between Fe2⁺ and HSA that promoted endocytosis. Moreover, the study elucidated that FcRn and caveolin regulated HSA endocytosis under normal conditions. Yet, in the presence of Fe2⁺, endocytosis shifted toward clathrin- and caveolin-enriched membrane domains, requiring both divalent metal transporter 1 (DMT1) and FcRn for efficient process completion. Thereby a new pathway is discovered for Fe2+-dependent HSA endocytosis involving DMT1 and clathrin. This findings highlighted the crucial role of iron in enhancing HSA endocytosis and transport, establishing Fe2+ as a vital endocytic enhancer that required binding to HSA to substantially improve its uptake. This insight into the mechanism of HSA endocytosis not only expands the understanding of protein transport but also opens new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Weibo Zhang
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Chong Chen
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Ran Wang
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Yixuan Li
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Yinhua Zhu
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Jiazeng Sun
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Yongting Luo
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Junjie Luo
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Liang Zhao
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
- Food Laboratory of Zhongyuan, Luohe, 462000, China
| | - Yanan Sun
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
- Food Laboratory of Zhongyuan, Luohe, 462000, China
| | - Huiyuan Guo
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
- Food Laboratory of Zhongyuan, Luohe, 462000, China
| | - Hao Zhang
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
- Food Laboratory of Zhongyuan, Luohe, 462000, China
| | - Bing Fang
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Yao Hu
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Rong Liu
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Xiaoyu Wang
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Jiayue Guo
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Ping Liu
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Peng An
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Pengjie Wang
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
- Food Laboratory of Zhongyuan, Luohe, 462000, China
| | - Juan Chen
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Fazheng Ren
- Co-constructed by Ministry of Education and Beijing Government, Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| |
Collapse
|
3
|
Fang B, Bai H, Zhang J, Shi M, Ge Y, Wang L, Li P, Ding Y, Zhang S, Zhang C, Qu Y, Zhang D, Peng B, Chen X, Li L, Huang W. Fluorogen-Activating Human Serum Albumin for Mitochondrial Nanoscale Imaging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2501849. [PMID: 40420673 DOI: 10.1002/adma.202501849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 05/16/2025] [Indexed: 05/28/2025]
Abstract
Fluorescence nanoscopy of living cells employs contrast agents to reveal intrinsic correlations between mitochondrial dynamics and functions at the molecular level. However, regular mitochondrial fluorophores usually present poor photostability, low brightness, non-specific inhibitory effects, high phototoxicity, and rapid photobleaching, which have hindered the use of these tools to capture the intricate dynamic features of mitochondria. Herein, we engineered a fluorogen-activating protein (FAP), AmpHecy@HSA, a non-covalent self-assembly of HSA and amphiphilic hemicyanine (AmpHecy) fluorophore, with exceptional cell permeability, long-lasting photostability, high brightness/fluorogenicity, and minimal phototoxicity. Crystallography and femtosecond transient absorption spectroscopy techniques were combined to elucidate the structural and mechanistic intricacies of fluorescence activation. These findings revealed that fluorophore photoactivation happens through the molecular conformation-induced intramolecular charge transfer, whose kinetics is mainly determined by the hydrophobic interaction between the fluorophore and nearby amino acids. This aligns with classical molecular dynamics simulations and excited-state conformation quantum mechanics. It was further demonstrated that AmpHecy@HSA can be used for super-resolved images of mitochondria within living cells without apparent phototoxicity. This work expands the fluorescent toolkit based on FAP engineering for studying live-cell mitochondrial morphology and function, advancing the fields of chemistry and biomedicine.
Collapse
Affiliation(s)
- Bin Fang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Xiamen University, Xiamen, 361102, China
- Future Display Institute in Xiamen, Xiamen, 361005, China
| | - Hua Bai
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jiaxin Zhang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Mengwen Shi
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China
| | - Yihao Ge
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China
| | - Limin Wang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Panpan Li
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Yang Ding
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Shiji Zhang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Xiamen University, Xiamen, 361102, China
| | - Congcong Zhang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Xiamen University, Xiamen, 361102, China
| | - Yunwei Qu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Xiamen University, Xiamen, 361102, China
| | - Duoteng Zhang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Xiamen University, Xiamen, 361102, China
| | - Bo Peng
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xi Chen
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China
| | - Lin Li
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Xiamen University, Xiamen, 361102, China
- Future Display Institute in Xiamen, Xiamen, 361005, China
| | - Wei Huang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Xiamen University, Xiamen, 361102, China
- Future Display Institute in Xiamen, Xiamen, 361005, China
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| |
Collapse
|
4
|
Li Y, Lu Y, Lu W, Zhong N, Li N, Xu Z, Zhang J, Sun H, Wu F, Teng Z, Tang Y, Wang S. Transformable albumin-based nanocapsules selectively amplify tumor starvation and disulfidptosis through metabolic deception. J Control Release 2025; 383:113739. [PMID: 40258477 DOI: 10.1016/j.jconrel.2025.113739] [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: 02/16/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 04/23/2025]
Abstract
Starvation-based therapy has emerged as a promising approach for cancer treatment. However, tumors can effectively circumvent nutrient deprivation by enhancing the uptake of alternative nutrients such as albumin to attenuate the efficacy of starvation-inducing drugs. In this study, we aimed to exploit the compensatory ability of tumors for alternative nutrients to improve the selectivity of starvation therapy. Albumin nanocapsules were coupled with glucose oxidase (GOX) and loaded with V9302 to obtain nutrient-mimicking transformable nanocapsules (HGV) that induced glucose and glutamine depletion. The HGV entered tumors efficiently owing to their transformability and induced starvation, which in turn upregulated the albumin uptake of the tumors to further increase the internalization of nanocapsules as a positive feedback loop. This amplified starvation led to a significant accumulation of intracellular disulfides and triggered disulfidptosis in the tumor cells, which not only effectively inhibited the growth of primary tumors but also stimulated antitumor immune responses. Furthermore, the tumor selectivity of HGV reduced the hepatotoxicity of GOX and V9302, making it a potential translational starvation-inducing nanocapsule.
Collapse
Affiliation(s)
- Yang Li
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yishi Lu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Wei Lu
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Nan Zhong
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Na Li
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Ziqing Xu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jie Zhang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Hanyao Sun
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Feiyun Wu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, China.
| | - Yuxia Tang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| | - Shouju Wang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| |
Collapse
|
5
|
Baldan M, Zhang S, Sun Q, Su Y, Mei D, Sun R, Zheng A, Liu D, Zhang J, Huo R, Tian Y, Han L, Wang S, Wang Y, Cui C. NIR-Triggered siRNA Release and Lysosomal Escape for Synergistic Photothermal Tumor Therapy. Int J Nanomedicine 2025; 20:4863-4882. [PMID: 40259913 PMCID: PMC12010081 DOI: 10.2147/ijn.s511655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/26/2025] [Indexed: 04/23/2025] Open
Abstract
Background Resistance to traditional treatments has spurred research into innovative therapeutic approaches for tumors. Among these innovative treatments, photothermal therapy (PTT) has gained increasing attention for its use of photothermal agents (PTAs) to convert light into heat for localized tumor ablation. However, PTT faces limitations due to heat shock protein 70 (HSP70)-mediated resistance in tumor cells. Combining PTT via indocyanine green (ICG) with siRNA HSP70 could reduce the thermal resistance of the tumor, thereby enhancing treatment efficacy. Albumin-based nanoparticles (NPs) can effectively deliver ICG and siRNA into tumor cells. When exposed to near-infrared (NIR) light, these nanoparticles trigger lysosomal escape and release, further enhancing gene silencing activity. Methods This study aimed to develop a biocompatible delivery system, HSA@ICG/siRNA NPs, for photothermal-enhanced tumor therapy. The nanoparticles were characterized for size, charge, surface functionalization, and photoconversion properties. In vitro antitumor efficacy was evaluated using MTT assay, calcein AM/PI staining, RT-PCR, and Western blot in 4T1 tumor cells. In vivo, we assessed photothermal effects, biodistribution, tumor inhibition, and biosafety following irradiation. Results Characterization confirmed the successful synthesis of uniform, stable HSA@ICG/siRNA NPs with effective photothermal conversion properties. Cellular uptake studies revealed high siRNA internalization, with laser-induced lysosomal escape enhancing cytoplasmic delivery. In vitro, gene silencing reduced mRNA and protein levels by 82.8% and 65%, respectively. In vivo, local tumor temperature increased to 42°C within 3 minutes, indicating a mild but effective photothermal effect. Tumor inhibition rates were 50.00% ± 9.16% for HSA@ICG and 71.26% ± 7.92% for HSA@ICG/siRNA, demonstrating enhanced tumor suppression. The treatment achieved sustained tumor targeting with minimal off-target toxicity. Conclusion As a dual-function photothermal therapy agent, HSA@ICG/siRNA NPs combine targeted gene silencing with photothermal effects, demonstrating significant therapeutic promise. This integrated approach addresses tumor resistance, offering a potential advancement in cancer treatment strategies.
Collapse
Affiliation(s)
- Myagmarsuren Baldan
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Shuang Zhang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Qi Sun
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Yan Su
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Dong Mei
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
- Department of Pharmacy, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Ran Sun
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Ao Zheng
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Danni Liu
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Jie Zhang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Ran Huo
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Yang Tian
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Le Han
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Shibo Wang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Yaoqi Wang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Chunying Cui
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Hu H, Li C, Song Y, Xie J, Li Q, Ke F, Wen B, Wang S, Gao W, Sun D. Albumin nanocomplex of BCL-2/xL inhibitor reduced platelet toxicity and improved anticancer efficacy in myeloproliferative neoplasm and lymphoma. Biomaterials 2025; 322:123347. [PMID: 40306157 DOI: 10.1016/j.biomaterials.2025.123347] [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: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 05/02/2025]
Abstract
The clinical application of BCL-2/xL inhibitors for cancer treatment is limited by the on-target thrombocytopenia. Although APG-1252 was designed to mitigate this issue, platelet toxicity at higher doses in clinical trials restricts dose escalation for greater efficacy. We have developed albumin nanocomplexes of APG-1252 (Nano-1252) to reduce platelet toxicity while improving drug efficacy through enhancing drug delivery to lymphoid organs. Nano-1252 forms stable nanoparticles due to the strong binding affinity between APG-1252 and albumin, reducing the platelet toxicity threshold by fourfold by limiting premature drug release and conversion to its active forms in circulation. Furthermore, Nano-1252 exhibited preferential accumulation in lymphoid organs, leading to enhanced anticancer efficacy in Mantle Cell Lymphoma (MCL) and Myeloproliferative Neoplasms (MPNs) mouse models. Our study not only develops a potential formulation to overcome the current translational barrier of APG-1252 but also reveals novel properties of the well-established albumin nanoformulation, thereby expanding its clinical applications.
Collapse
Affiliation(s)
- Hongxiang Hu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chengyi Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yudong Song
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jizhao Xie
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Qiuxia Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Fang Ke
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shaomeng Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Wei Gao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Pharmacology and Pharmaceutical Science, College of Pharmacy, The University of Houston, TX, 77204, USA.
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
7
|
Xu G, Zhang Q, Cheng R, Qu J, Li W. Survival strategies of cancer cells: the role of macropinocytosis in nutrient acquisition, metabolic reprogramming, and therapeutic targeting. Autophagy 2025; 21:693-718. [PMID: 39817564 PMCID: PMC11925119 DOI: 10.1080/15548627.2025.2452149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
Macropinocytosis is a nonselective form of endocytosis that allows cancer cells to largely take up the extracellular fluid and its contents, including nutrients, growth factors, etc. We first elaborate meticulously on the process of macropinocytosis. Only by thoroughly understanding this entire process can we devise targeted strategies against it. We then focus on the central role of the MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1) in regulating macropinocytosis, highlighting its significance as a key signaling hub where various pathways converge to control nutrient uptake and metabolic processes. The article covers a comprehensive analysis of the literature on the molecular mechanisms governing macropinocytosis, including the initiation, maturation, and recycling of macropinosomes, with an emphasis on how these processes are hijacked by cancer cells to sustain their growth. Key discussions include the potential therapeutic strategies targeting macropinocytosis, such as enhancing drug delivery via this pathway, inhibiting macropinocytosis to starve cancer cells, blocking the degradation and recycling of macropinosomes, and inducing methuosis - a form of cell death triggered by excessive macropinocytosis. Targeting macropinocytosis represents a novel and innovative approach that could significantly advance the treatment of cancers that rely on this pathway for survival. Through continuous research and innovation, we look forward to developing more effective and safer anti-cancer therapies that will bring new hope to patients.Abbreviation: AMPK: AMP-activated protein kinase; ASOs: antisense oligonucleotides; CAD: carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase; DC: dendritic cell; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; ERBB2: erb-b2 receptor tyrosine kinase 2; ESCRT: endosomal sorting complex required for transport; GAP: GTPase-activating protein; GEF: guanine nucleotide exchange factor; GRB2: growth factor receptor bound protein 2; LPP: lipopolyplex; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; MTORC2: mechanistic target of rapamycin kinase complex 2; NSCLC: non-small cell lung cancer; PADC: pancreatic ductal adenocarcinoma; PDPK1: 3-phosphoinositide dependent protein kinase 1; PI3K: phosphoinositide 3-kinase; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; PtdIns(3,4,5)P3: phosphatidylinositol-(3,4,5)-trisphosphate; PtdIns(4,5)P2: phosphatidylinositol-(4,5)-bisphosphate; PTT: photothermal therapies; RAC1: Rac family small GTPase 1; RPS6: ribosomal protein S6; RPS6KB1: ribosomal protein S6 kinase B1; RTKs: receptor tyrosine kinases; SREBF: sterol regulatory element binding transcription factor; TFEB: transcription factor EB; TNBC: triple-negative breast cancer; TSC2: TSC complex subunit 2; ULK1: unc-51 like autophagy activating kinase 1; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Guoshuai Xu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Qinghong Zhang
- Emergency Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Renjia Cheng
- Department of Intensive Care Medicine, The General Hospital of the Northern Theater Command of the People’s Liberation Army of China, Shenyang, Liaoning, China
| | - Jun Qu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Wenqiang Li
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
8
|
Rong D, Gao L, Chen Y, Gao XZ, Tang M, Tang H, Gao Y, Lu G, Ling ZQ, Shen HM. Suppression of the LKB1-AMPK-SLC7A11-GSH signaling pathway sensitizes NSCLC to albumin-bound paclitaxel via oxidative stress. Redox Biol 2025; 81:103567. [PMID: 40023979 PMCID: PMC11915006 DOI: 10.1016/j.redox.2025.103567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Albumin-bound paclitaxel (nab-PTX) is an important chemotherapeutic drug used for the treatment of advanced and metastatic non-small cell lung cancer (NSCLC). One critical issue in its clinical application is the development of resistance; thus, a deeper understanding of the mechanisms underlying the primary resistance to nab-PTX is expected to help to develop effective therapeutic strategies to overcome resistance. In this study, we made an unexpected discovery that NSCLC with wild-type (WT) Liver kinase B1 (LKB1), an important tumor suppressor and upstream kinase of AMP-activated protein kinase (AMPK), is more resistant to nab-PTX than NSCLC with mutant LKB1. Mechanistically, LKB1 status does not alter the intracellular concentration of nab-PTX or affect its canonical pharmacological action in promoting microtubule polymerization. Instead, we found that LKB1 mediates AMPK activation, leading to increased expression of SLC7A11, a key amino acid transporter and intracellular level of glutathione (GSH), which then attenuates the production of reactive oxygen species (ROS) and apoptotic cell death induced by nab-PTX. On the other hand, genetic or pharmacological inhibition of AMPK in LKB1-WT NSCLC reduces the expression of SLC7A11 and intracellular GSH, increases ROS level, and eventually promotes the apoptotic cell death induced by nab-PTX in vitro. Consistently, the combination of nab-PTX with an AMPK inhibitor exhibits a greater therapeutic efficacy in LKB1-WT NSCLC using xenograft models in vivo. Taken together, our data reveal a novel role of LKB1-AMPK-SLC7A11-GSH signaling pathway in the primary resistance to nab-PTX, and provide a therapeutic strategy for the treatment of LKB1-WT NSCLC by targeting the LKB1-AMPK-SLC7A11-GSH pathway.
Collapse
Affiliation(s)
- Dade Rong
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Liangliang Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Yiguan Chen
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Xiang-Zheng Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Mingzhu Tang
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Haimei Tang
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China; Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Yuan Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Guang Lu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Qiang Ling
- Experimental Research Centre, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Han-Ming Shen
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China.
| |
Collapse
|
9
|
Sun M, He L, Chen R, Lv M, Chen ZS, Fan Z, Zhou Y, Qin J, Du J. Rational design of peptides to overcome drug resistance by metabolic regulation. Drug Resist Updat 2025; 79:101208. [PMID: 39914188 DOI: 10.1016/j.drup.2025.101208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/24/2025]
Abstract
Chemotherapy is widely used clinically, however, its efficacy is often compromised by the development of drug resistance, which arises from prolonged administration of drugs or other stimuli. One of the driven causes of drug resistance in tumors or bacterial infections is metabolic reprogramming, which alters mitochondrial metabolism, disrupts metabolic pathways and causes ion imbalance. Bioactive peptide materials, due to their biocompatibility, diverse bioactivities, customizable sequences, and ease of modification, have shown promise in overcoming drug resistance. This review provides an in-depth analysis of metabolic reprogramming and associated microenvironmental changes that contribute to drug resistance in common tumors and bacterial infections, suggesting potential therapeutic targets. Additionally, we explore peptide-based materials for regulating metabolism and their potential synergic effect with other therapies, highlighting the mechanisms by which these peptides reverse drug resistance. Finally, we discuss future perspectives and the clinical challenges in peptide-based treatments, aiming to offer insights for overcoming drug-resistant diseases.
Collapse
Affiliation(s)
- Min Sun
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Le He
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Ran Chen
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Mingchen Lv
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Zhen Fan
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yuxiao Zhou
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
| | - Jinlong Qin
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China.
| | - Jianzhong Du
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China; Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China.
| |
Collapse
|
10
|
Liu S, Liu Z, Lei H, Miao YB, Chen J. Programmable Nanomodulators for Precision Therapy, Engineering Tumor Metabolism to Enhance Therapeutic Efficacy. Adv Healthc Mater 2025; 14:e2403019. [PMID: 39529548 DOI: 10.1002/adhm.202403019] [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: 08/13/2024] [Revised: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Tumor metabolism is crucial in the continuous advancement and complex growth of cancer. The emerging field of nanotechnology has made significant strides in enhancing the understanding of the complex metabolic intricacies inherent to tumors, offering potential avenues for their strategic manipulation to achieve therapeutic goals. This comprehensive review delves into the interplay between tumor metabolism and various facets of cancer, encompassing its origins, progression, and the formidable challenges posed by metastasis. Simultaneously, it underscores the classification of programmable nanomodulators and their transformative impact on enhancing cancer treatment, particularly when integrated with modalities such as chemotherapy, radiotherapy, and immunotherapy. This review also encapsulates the mechanisms by which nanomodulators modulate tumor metabolism, including the delivery of metabolic inhibitors, regulation of oxidative stress, pH value modulation, nanoenzyme catalysis, nutrient deprivation, and RNA interference technology, among others. Additionally, the review delves into the prospects and challenges of nanomodulators in clinical applications. Finally, the innovative concept of using nanomodulators to reprogram metabolic pathways is introduced, aiming to transform cancer cells back into normal cells. This review underscores the profound impact that tailored nanomodulators can have on tumor metabolic, charting a path toward pioneering precision therapies for cancer.
Collapse
Affiliation(s)
- Siwei Liu
- Women & Children's Molecular Medicine Center, Department of Gynecology, Guangyuan Central Hospital, No. 16, Jingxiangzi, Lizhou District, Guangyuan, 628000, P. R. China
| | - Zhijun Liu
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China
| | - Huajiang Lei
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Jiao Chen
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| |
Collapse
|
11
|
Haanen TJ, Boock S, Callahan CG, Peris I, Zawacki KP, Raines B, Nino CA, Tran B, Harold A, Onishi GH, Hinderman M, Dowdican A, Huang W, Taylor DJ, Taylor SE, Jackson MW, DiFeo A, O’Connor CM, Narla G. Mutant PP2A Induces IGFBP2 Secretion to Promote Development of High-Grade Uterine Cancer. Cancer Res 2025; 85:442-461. [PMID: 39531506 PMCID: PMC11788061 DOI: 10.1158/0008-5472.can-24-1263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/09/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Uterine serous carcinoma (USC) and uterine carcinosarcoma (UCS) tumors are uniquely aggressive, suggesting that the primary tumor is intrinsically equipped to disseminate and metastasize. Previous work identified mutational hotspots within PPP2R1A, which encodes the Aα scaffolding subunit of protein phosphatase 2A (PP2A), a heterotrimeric serine/threonine phosphatase. Two recurrent heterozygous PPP2R1A mutations, P179R and S256F, occur exclusively within high-grade subtypes of uterine cancer and can drive tumorigenesis and metastasis. Elucidation of the mechanisms by which PP2A Aα mutants promote tumor development and progression could help identify therapeutic opportunities. Here, we showed that expression of these mutants in USC/UCS cell lines enhanced tumor-initiating capacity, drove a hybrid epithelial-to-mesenchymal plasticity phenotype, and elevated secretion of the tumorigenic cytokine insulin growth factor (IGF) binding protein 2 (IGFBP2). Therapeutic targeting of the IGFBP2/IGF receptor 1 signaling axis using small molecules and genetic approaches resulted in marked tumor growth inhibition. Mechanistically, PP2A regulated IGFBP2 expression through the transcription factor, NF-κB, which harbors a B56 recognition motif. Collectively, these results identify a role for PP2A in regulating paracrine cancer cell signaling that can be targeted to block the initiation and metastasis of high-grade uterine cancer. Significance: Elevated IGFBP2 secretion by uterine cancer cells with heterozygous PPP2R1A mutations supports tumor progression and confers a vulnerability to IGFBP2/IGF1R inhibition as a therapeutic approach for this highly aggressive cancer subtype.
Collapse
Affiliation(s)
- Terrance J. Haanen
- Department of Cancer Biology, The University of Michigan, Ann Arbor, MI
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
| | - Sophie Boock
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
| | - Catherine G. Callahan
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
| | - Irene Peris
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
| | - Kaitlin P. Zawacki
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
| | - Brynne Raines
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
- Department of Cellular and Molecular Biology, The University of Michigan, Ann Arbor, MI
| | - Charles A. Nino
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
- Department of Cellular and Molecular Biology, The University of Michigan, Ann Arbor, MI
| | - Brian Tran
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
- Department of Pharmacology, The University of Michigan, Ann Arbor, MI
| | - Alexis Harold
- Department of Cancer Biology, The University of Michigan, Ann Arbor, MI
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
| | - Gabrielle Hodges Onishi
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
| | - Matthew Hinderman
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
| | - Amanda Dowdican
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
| | - Wei Huang
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH
| | - Derek J. Taylor
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH
| | - Sarah E. Taylor
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Mark W. Jackson
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Analisa DiFeo
- Rogel Cancer Center, The University of Michigan Health, Ann Arbor, Michigan
- Department of Pathology, The University of Michigan, Ann Arbor, Michigan
- Department of Obstetrics and Gynecology, The University of Michigan, Ann Arbor, Michigan
| | - Caitlin M. O’Connor
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
- Rogel Cancer Center, The University of Michigan Health, Ann Arbor, Michigan
| | - Goutham Narla
- Department of Internal Medicine: Division of Genetic Medicine, The University of Michigan, Ann Arbor, MI
- Rogel Cancer Center, The University of Michigan Health, Ann Arbor, Michigan
| |
Collapse
|
12
|
Tang Y, Yu X, He L, Tang M, Yue W, Chen R, Zhao J, Pan Q, Li W. A high-valence bismuth(V) nanoplatform triggers cancer cell death and anti-tumor immune responses with exogenous excitation-free endogenous H 2O 2- and O 2-independent ROS generation. Nat Commun 2025; 16:860. [PMID: 39833161 PMCID: PMC11747550 DOI: 10.1038/s41467-025-56110-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Reactive oxygen species with evoked immunotherapy holds tremendous promise for cancer treatment but has limitations due to its dependence on exogenous excitation and/or endogenous H2O2 and O2. Here we report a versatile oxidizing pentavalent bismuth(V) nanoplatform (NaBiVO3-PEG) can generate reactive oxygen species in an excitation-free and H2O2- and O2-independent manner. Upon exposure to the tumor microenvironment, NaBiVO3-PEG undergoes continuous H+-accelerated hydrolysis with •OH and 1O2 generation through electron transfer-mediated BiV-to-BiIII conversion and lattice oxygen transformation. The simultaneous release of sodium counterions after endocytosis triggers caspase-1-mediated pyroptosis. NaBiVO3-PEG intratumorally administered initiates robust therapeutic efficacies against both primary and distant tumors and activates systemic immune responses to combat tumor metastasis. NaBiVO3-PEG intravenously administered can efficiently accumulate at the tumor site for further real-time computed tomography monitoring, immunotherapy, or alternative synergistic immune-radiotherapy. Overall, this work offers a nanomedicine based on high-valence bismuth(V) nanoplatform and underscores its great potential for cancer immunotherapy.
Collapse
Affiliation(s)
- Yizhang Tang
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering Shanghai Jiao Tong University 800 Dongchuan Road, Shanghai, P. R. China
- Future Material Innovation Center Zhangjiang Institute for Advanced Study Shanghai Jiao Tong University 429 Zhangheng Road, Shanghai, P. R. China
| | - Xujiang Yu
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering Shanghai Jiao Tong University 800 Dongchuan Road, Shanghai, P. R. China.
- Future Material Innovation Center Zhangjiang Institute for Advanced Study Shanghai Jiao Tong University 429 Zhangheng Road, Shanghai, P. R. China.
| | - Liangrui He
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering Shanghai Jiao Tong University 800 Dongchuan Road, Shanghai, P. R. China
| | - Meng Tang
- Department of Comprehensive Oncology National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College 17 Panjiayuan South Lane, Beijing, P. R. China
| | - Wenji Yue
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering Shanghai Jiao Tong University 800 Dongchuan Road, Shanghai, P. R. China
| | - Ruitong Chen
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering Shanghai Jiao Tong University 800 Dongchuan Road, Shanghai, P. R. China
| | - Jie Zhao
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering Shanghai Jiao Tong University 800 Dongchuan Road, Shanghai, P. R. China
| | - Qi Pan
- Department of Urology Shanghai General Hospital Shanghai Jiao Tong University School of Medicine 85 Wujin Road, Shanghai, P. R. China
| | - Wanwan Li
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering Shanghai Jiao Tong University 800 Dongchuan Road, Shanghai, P. R. China.
- Future Material Innovation Center Zhangjiang Institute for Advanced Study Shanghai Jiao Tong University 429 Zhangheng Road, Shanghai, P. R. China.
| |
Collapse
|
13
|
Wang X, Yu G. Advancing veterinary vaccines design through trained immunity insights. Front Vet Sci 2025; 11:1524668. [PMID: 39881716 PMCID: PMC11776093 DOI: 10.3389/fvets.2024.1524668] [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: 11/08/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Trained immunity, characterized by long-term functional reprogramming of innate immune cells, offers promising new directions for veterinary vaccine development. This perspective examines how trained immunity can be integrated into veterinary vaccine design through metabolic reprogramming and epigenetic modifications. We analyze key molecular mechanisms, including the shift to aerobic glycolysis and sustained epigenetic changes, that enable enhanced immune responses. Strategic approaches for vaccine optimization are proposed, focusing on selecting effective trained immunity inducers, developing innovative adjuvant systems, and achieving synergistic enhancement of immune responses. While implementation challenges exist, including individual response variations and safety considerations, trained immunity-based vaccines show potential for providing broader protection against emerging pathogens. This approach could revolutionize veterinary vaccinology by offering enhanced efficacy and cross-protection against heterologous infections, particularly valuable for zoonotic disease control.
Collapse
Affiliation(s)
- Xin Wang
- College of Life Science, Longyan University, Longyan, China
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnolog, Longyan, China
- Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, China
- Chinese International College, Dhurakij Pundit University, Bangkok, Thailand
| | - Guohua Yu
- College of Life Science, Longyan University, Longyan, China
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnolog, Longyan, China
- Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, China
| |
Collapse
|
14
|
Ren Y, Li P, Xie Y, Xu J, Luo Q, Chen M, Liu R, Feng H, Chen Y, Liu Y, Bao C, Duan J, Li J, Lu W. Dual-responsive nanoparticles for enhanced drug delivery in breast Cancer chemotherapy. J Control Release 2025; 377:146-161. [PMID: 39549730 DOI: 10.1016/j.jconrel.2024.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
Drug delivery efficiency often affects chemotherapy outcome due to dense collagen barrier in tumor environment. Here, we report a nanoparticle capable of pH and glutathione dual-responsive drug delivery to enhance the efficacy of breast cancer chemotherapy. Maleiminated polyethylene glycol and polylactide block copolymer were synthesized as a core material, doxorubicin was encapsulated into the nanoparticle by self-assembly. Thiocollagenase and maleimide were connected on the nanoparticle surface by click chemistry, and further coated with chondroitin sulfate as a protective layer to form dual-responsive doxorubicin nanoparticle. The results showed that the nanoparticle had the ability to penetrate deep tumor tissue, to target on CD44 of cancer cell, and to release doxorubicin in cancer cell in response to pH and glutathione signals, demonstrating superior anticancer efficacy in breast cancer-bearing mice. In conclusion, the dual-responsive nanoparticle could be used as a drug carrier to enhance drug delivery in breast cancer chemotherapy.
Collapse
Affiliation(s)
- Yuxin Ren
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Peishan Li
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ying Xie
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jiarui Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Qian Luo
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ming Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Rui Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Hexuan Feng
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yuling Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yixuan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Chunjie Bao
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jialun Duan
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jianwei Li
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China; School of Pharmacy, Changzhi Medical College, Changzhi 046000, China; Beijing Zhendong Guangming Pharmaceutical Research Institute Co., Ltd, Beijing 100080, China
| | - Wanliang Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing, China.
| |
Collapse
|
15
|
Soni SS, Rodell CB. Cyclodextrin Nanoparticles and Injectable Polymer-Nanoparticle Hydrogels for Macrophage-Targeted Delivery of Small-Molecule Drugs. Methods Mol Biol 2025; 2902:117-131. [PMID: 40029599 DOI: 10.1007/978-1-0716-4402-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Small-molecule drugs are pillars of modern medicine, used to prevent and treat a plethora of diseases. Their applications include modulating the immune response, where macrophages and other phagocytic innate immune cells are critical therapeutic targets. In comparison to biologics, small-molecule drugs benefit from relative ease of synthesis and structural modification, long shelf life, and low cost. However, these drugs often suffer from poor pharmacokinetics (i.e., rapid renal clearance, nonspecific tissue, and cell biodistribution) and off-target effects that limit their efficacy. We have, therefore, developed a macrophage-targeted nanoparticulate carrier (cyclodextrin nanoparticle [CDNP]) that can house a variety of hydrophobic small-molecule drugs for systemic delivery through the body. This system has further been manipulated to formulate an injectable polymer-nanoparticle (iPNP) hydrogel for local delivery, with the goal of concentrating drug effects at the target site while perpetuating sustained delivery and minimizing off-target effects. Herein, we describe the strategy for preparing these CDNPs and iPNP hydrogels for a wide range of therapeutic applications.
Collapse
Affiliation(s)
- Shreya S Soni
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Christopher B Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Wang J, Yu N, Tang Y, Cheng Y, Li H. FDA-Approved Hydrogel-Mediated In Situ Sonodynamic and Chemotherapeutic Therapy for Pancreatic Cancer. Pharmaceuticals (Basel) 2024; 17:1666. [PMID: 39770508 PMCID: PMC11678859 DOI: 10.3390/ph17121666] [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: 11/06/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Albumin-bound paclitaxel (nab-PTX) nanoparticles have been proven effective in treating advanced pancreatic cancer. However, the clinical application of nab-PTX nanoparticles is often associated with suboptimal outcomes and severe side effects due to its non-specific distribution and rapid clearance. This study aims to develop a novel nanoplatform that integrates sonodynamic therapy (SDT) and chemotherapy to enhance treatment efficacy and reduce systemic side effects. Methods: Bovine serum albumin (BSA) was conjugated with chlorin e6 and paclitaxel (PTX) to form stable nanoparticles (NPs). These NPs were then incorporated into a biodegradable poly(lactic-co-glycolic acid)-b-polyethylene glycol-b-poly(lactic-co-glycolic acid) hydrogel for targeted drug delivery. The system's stability and drug release profile were analyzed, followed by in vitro studies to evaluate cellular uptake and cancer cell killing efficacy. In vivo evaluation was performed using pancreatic cancer xenograft models, with intratumoral injection of the drug-loaded hydrogel. Results: The developed hydrogel system demonstrated enhanced stability and sustained release of PTX. In vitro analyses revealed significant cellular uptake and synergistic cancer cell killing effects through combined SDT and chemotherapy. In vivo studies showed prolonged intratumoral retention of the drug and remarkable inhibition of tumor growth. Conclusions: This novel nanoplatform offers a promising approach for improving pancreatic cancer treatment by enhancing intratumoral drug retention and minimizing systemic side effects. The synergistic effects of SDT and chemotherapy demonstrate the potential of this strategy in achieving better therapeutic outcomes.
Collapse
Affiliation(s)
- Jian Wang
- Department of Radiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Nianhui Yu
- Department of Radiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Yunpeng Tang
- Department of Radiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200231, China
| | - Yingsheng Cheng
- Department of Imaging Medicine and Nuclear Medicine, Tongji Hospital, Shanghai 200065, China
| | - Hui Li
- Department of Radiology, Sixth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
17
|
Zhu S, Wu Q, Ying Y, Mao Y, Lu W, Xu J, Cai X, He H, Wu J. Tissue-Adaptive BSA Hydrogel with Dual Release of PTX and bFGF Promotes Spinal Cord Injury Repair via Glial Scar Inhibition and Axon Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401407. [PMID: 39385643 DOI: 10.1002/smll.202401407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/18/2024] [Indexed: 10/12/2024]
Abstract
Spinal cord injury (SCI) is a severe clinical disease usually accompanied by activated glial scar, neuronal axon rupture, and disabled motor function. To mimic the microenvironment of the SCI injury site, a hydrogel system with a comparable mechanical property to the spinal cord is desirable. Therefore, a novel elastic bovine serum albumin (BSA) hydrogel is fabricated with excellent adhesive, injectable, and biocompatible properties. The hydrogel is used to deliver paclitaxel (PTX) together with basic fibroblast growth factor (bFGF) to inhibit glial scar formation as well as promote axon regeneration and motor function for SCI repair. Due to the specific interaction of BSA with both drugs, bFGF, and PTX can be controllably released from the hydrogel system to achieve an effective concentration at the wound site during the SCI regeneration process. Moreover, benefiting from the combination of PTX and bFGF, this bFGF/PTX@BSA system significantly aided axon repair by promoting the elongation of axons across the glial scar with reduced reactive astrocyte secretion. In addition, remarkable anti-apoptosis of nerve cells is evident with the bFGF/PTX@BSA system. Subsequently, this multi-functionalized drug system significantly improved the motor function of the rats after SCI. These results reveal that bFGF/PTX@BSA is an ideal functionalized material for nerve repair in SCI.
Collapse
Affiliation(s)
- Sipin Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Qiuji Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yibo Ying
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yuqin Mao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wenjie Lu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jie Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiong Cai
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Huacheng He
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Jiang Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
18
|
Chen Y, Xu W, Jin H, Zhang M, Liu S, Liu Y, Zhang H. Nutritional Glutamine-Modified Iron-Delivery System with Enhanced Endocytosis for Ferroptosis Therapy of Pancreatic Tumors. ACS NANO 2024; 18:31846-31868. [PMID: 39512234 DOI: 10.1021/acsnano.4c08083] [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: 11/15/2024]
Abstract
Heterogeneous reprogrammed nutrient metabolic networks formed by oncogenes exhibit the potential for exploring novel druggable targets and developing innovative anticancer therapeutics. Herein, based on the heterogeneous metabolic characteristics of glutamine (Gln) addiction in pancreatic cancer cells, an iron-delivery system (IDS) with enhanced endocytosis is designed for efficient ferroptosis therapy. The IDS is characterized by Gln modification and can be recognized as a source of Gln nutrients for efficient endocytic uptake by pancreatic tumor cells. Because the IDS is flexible to combine with amino acid-like components, the IDS with enhanced endocytosis is further produced by loading the Gln transporter inhibitor of V9302. V9302 is capable of suppressing molecular Gln uptake via transporter ASCT2, which generates Gln deprivation to direct metabolic reprogramming of cancer cells and enhances cellular uptake of Gln-modified IDS via RAS-stimulated macropinocytosis. The enhanced endocytosis and high iron content of IDS facilitate ferroptosis in mice pancreatic tumor models; thus, an amino acid-like ferroptosis inducer of l-buthionine sulfoximine (BSO) is further combined. The enhanced endocytosis resulting from the synergism of Gln and V9302 enables the efficient delivery of iron and BSO for ferroptosis tumor therapy. This work provides an alternative approach for enhancing intracellular drug delivery of the tumors with heterogeneous nutrient metabolism by virtue of combining nutrient-modified nanodrugs with the corresponding nutrient transporter inhibitors.
Collapse
Affiliation(s)
- Yang Chen
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Wenzhe Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Hao Jin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Mengsi Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Shuwei Liu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Yi Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, P. R. China
| |
Collapse
|
19
|
Song J, Cho MH, Cho H, Song Y, Lee SW, Nam HC, Yoon TH, Shin JC, Hong JS, Kim Y, Ekanayake E, Jeon J, You DG, Im SG, Choi GS, Park JS, Carter BC, Balaj L, Seo AN, Miller MA, Park SY, Kang T, Castro CM, Lee H. Amplifying mutational profiling of extracellular vesicle mRNA with SCOPE. Nat Biotechnol 2024:10.1038/s41587-024-02426-6. [PMID: 39375445 DOI: 10.1038/s41587-024-02426-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/09/2024] [Indexed: 10/09/2024]
Abstract
Sequencing of messenger RNA (mRNA) found in extracellular vesicles (EVs) in liquid biopsies can provide clinical information such as somatic mutations, resistance profiles and tumor recurrence. Despite this, EV mRNA remains underused due to its low abundance in liquid biopsies, and large sample volumes or specialized techniques for analysis are required. Here we introduce Self-amplified and CRISPR-aided Operation to Profile EVs (SCOPE), a platform for EV mRNA detection. SCOPE leverages CRISPR-mediated recognition of target RNA using Cas13 to initiate replication and signal amplification, achieving a sub-attomolar detection limit while maintaining single-nucleotide resolution. As a proof of concept, we designed probes for key mutations in KRAS, BRAF, EGFR and IDH1 genes, optimized protocols for single-pot assays and implemented an automated device for multi-sample detection. We validated SCOPE's ability to detect early-stage lung cancer in animal models, monitored tumor mutational burden in patients with colorectal cancer and stratified patients with glioblastoma. SCOPE can expedite readouts, augmenting the clinical use of EVs in precision oncology.
Collapse
Affiliation(s)
- Jayeon Song
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Forensic Sciences, Sungkyunkwan University, Suwon, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Mi Hyeon Cho
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hayoung Cho
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Younseong Song
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | | | - Ho Chul Nam
- RevoSketch, Inc., Daejeon, Republic of Korea
| | - Tae Ho Yoon
- RevoSketch, Inc., Daejeon, Republic of Korea
| | | | - Jae-Sang Hong
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yejin Kim
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Emil Ekanayake
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Jueun Jeon
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dong Gil You
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Gyu-Seog Choi
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jun Seok Park
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Bob C Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - An Na Seo
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Soo Yeun Park
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Taejoon Kang
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.
| | - Cesar M Castro
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA.
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA.
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
| |
Collapse
|
20
|
Gong Z, Fu Y, Gao Y, Jiao F, Su Q, Sang X, Chen B, Deng X, Liu X. "Abraxane-Like" Radiosensitizer for In Situ Oral Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309569. [PMID: 38973195 PMCID: PMC11425904 DOI: 10.1002/advs.202309569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 06/06/2024] [Indexed: 07/09/2024]
Abstract
Radiotherapy plays a vital role in cancer therapy. However, the hypoxic microenvironment of tumors greatly limits the effectiveness, thus it is crucial to develop a simple, efficient, and safe radiosensitizer to reverse hypoxia and ameliorate the efficacy of radiotherapy. Inspired by the structure of canonical nanodrug Abraxane, herein, a native HSA-modified CaO2 nanoparticle system (CaO2-HSA) prepared by biomineralization-induced self-assembly is developed. CaO2-HSA will accumulate in tumor tissue and decompose to produce oxygen, altering the hypoxic condition inside the tumor. Simultaneously, ROS and calcium ions will lead to calcium overload and further trigger immunogenic cell death. Notably, its sensitizing enhancement ratio (SER = 3.47) is much higher than that of sodium glycididazole used in the clinic. Furthermore, in animal models of in situ oral cancer, CaO2-HSA can effectively inhibit tumor growth. With its high efficacy, facile preparation, and heavy-metal free biosafety, the CaO2-HSA-based radiosensitizer holds enormous potential for oral cancer therapy.
Collapse
Affiliation(s)
- Zijian Gong
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yixuan Fu
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yuan Gao
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Fei Jiao
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Qinzhi Su
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xiao Sang
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Binglin Chen
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xuliang Deng
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Biomedical Engineering DepartmentPeking UniversityBeijing100191P. R. China
| | - Xinyu Liu
- Central LaboratoryDepartment of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and MaterialTechnology of StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| |
Collapse
|
21
|
Soni S, Kim KM, Sarkar B, Rodell CB. Uptake of Cyclodextrin Nanoparticles by Macrophages is Dependent on Particle Size and Receptor-Mediated Interactions. ACS APPLIED BIO MATERIALS 2024; 7:4856-4866. [PMID: 38231485 PMCID: PMC11252246 DOI: 10.1021/acsabm.3c00985] [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: 10/22/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/18/2024]
Abstract
Physiochemical properties of nanoparticles, such as their size and chemical composition, dictate their interaction with professional phagocytes of the innate immune system. Macrophages, in particular, are key regulators of the immune microenvironment that heavily influence particle biodistribution as a result of their uptake. This attribute enables macrophage-targeted delivery, including for phenotypic modulation. Saccharide-based materials, including polyglucose polymers and nanoparticles, are efficient vehicles for macrophage-targeted delivery. Here, we investigate the influence of particle size on cyclodextrin nanoparticle (CDNP) uptake by macrophages and further examine the receptor-mediated interactions that drive macrophage-targeted delivery. We designed and synthesized CDNPs ranging in size from 25 nm to >100 nm in diameter. Increasing particle size was correlated with greater uptake by macrophages in vitro. Both scavenger receptor A1 and mannose receptor were critical mediators of macrophage-targeted delivery, inhibition of which reduced the extent of uptake. Finally, we investigated the cellular bioavailability of drug-loaded CDNPs using a model anti-inflammatory drug, celastrol, which demonstrated that drug bioactivity is improved by CDNP loading relative to free drug alone. This study thus elucidates the interactions between the polyglucose nanoparticles and macrophages, thereby facilitating their application in macrophage-targeted drug delivery that has applications in the context of tissue injury and repair.
Collapse
Affiliation(s)
- Shreya
S. Soni
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Kenneth M. Kim
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
- Department
of Microbiology and Immunology, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Biplab Sarkar
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Christopher B. Rodell
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
22
|
Quintana JM, Jiang F, Kang M, Valladolid Onecha V, Könik A, Qin L, Rodriguez VE, Hu H, Borges N, Khurana I, Banla LI, Le Fur M, Caravan P, Schuemann J, Bertolet A, Weissleder R, Miller MA, Ng TSC. Localized in vivo prodrug activation using radionuclides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606075. [PMID: 39211146 PMCID: PMC11361159 DOI: 10.1101/2024.08.02.606075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Radionuclides used for imaging and therapy can show high molecular specificity in the body with appropriate targeting ligands. We hypothesized that local energy delivered by molecularly targeted radionuclides could chemically activate prodrugs at disease sites while avoiding activation in off-target sites of toxicity. As proof-of-principle, we tested whether this strategy of " RA dionuclide i nduced D rug E ngagement for R elease" ( RAiDER ) could locally deliver combined radiation and chemotherapy to maximize tumor cytotoxicity while minimizing exposure to activated chemotherapy in off-target sites. Methods We screened the ability of radionuclides to chemically activate a model radiation-activated prodrug consisting of the microtubule destabilizing monomethyl auristatin E caged by a radiation-responsive phenyl azide ("caged-MMAE") and interpreted experimental results using the radiobiology computational simulation suite TOPAS-nBio. RAiDER was evaluated in syngeneic mouse models of cancer using fibroblast activation protein inhibitor (FAPI) agents 99m Tc-FAPI-34 and 177 Lu-FAPI-04, the prostate-specific membrane antigen (PSMA) agent 177 Lu-PSMA-617, combined with caged-MMAE or caged-exatecan. Biodistribution in mice, combined with clinical dosimetry, estimated the relationship between radiopharmaceutical uptake in patients and anticipated concentrations of activated prodrug using RAiDER. Results RAiDER efficiency varied by 250-fold across radionuclides ( 99m Tc> 177 Lu> 64 Cu> 68 Ga> 223 Ra> 18 F), yielding up to 1.22µM prodrug activation per Gy of exposure from 99m Tc. Computational simulations implicated low-energy electron-mediated free radical formation as driving prodrug activation. Clinically relevant radionuclide concentrations chemically activated caged-MMAE restored its ability to destabilize microtubules and increased its cytotoxicity by up to 600-fold compared to non-irradiated prodrug. Mice treated with 99m Tc-FAPI-34 and caged-MMAE accumulated up to 3000× greater concentrations of activated MMAE in tumors compared to other tissues. RAiDER with 99m Tc-FAPI-34 or 177 Lu-FAPI-04 delayed tumor growth, while monotherapies did not ( P <0.03). Clinically-guided dosimetry suggests sufficient radiation doses can be delivered to activate therapeutically meaningful levels of prodrug. Conclusion This proof-of-concept study shows that RAiDER is compatible with multiple radionuclides commonly used in nuclear medicine and has the potential to improve the efficacy of radiopharmaceutical therapies to treat cancer safely. RAiDER thus shows promise as an effective strategy to treat disseminated malignancies and broadens the capability of radiopharmaceuticals to trigger diverse biological and therapeutic responses. Abstract Figure
Collapse
|
23
|
Qiu J, Jiang Y, Ye N, Jin G, Shi H, Qian D. Leveraging the intratumoral microbiota to treat human cancer: are engineered exosomes an effective strategy? J Transl Med 2024; 22:728. [PMID: 39103887 PMCID: PMC11302114 DOI: 10.1186/s12967-024-05531-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Cancer remains a leading cause of global mortality. The tumor microbiota has increasingly been recognized as a key regulator of cancer onset and progression, in addition to shaping tumor responses to immunotherapy. Microbes, including viruses, bacteria, fungi, and other eukaryotic species can impact the internal homeostasis and health of humans. Research focused on the gut microflora and the intratumoral microbiome has revolutionized the current understanding of how tumors grow, progress, and resist therapeutic interventions. Even with this research, however, there remains relatively little that is known with respect to the abundance of microbes and their effects on tumors and the tumor microenvironment. Engineered exosomes are a class of artificial extracellular nanovesicles that can actively transport small molecule drugs and nucleic acids, which have the broad prospects of tumor cell therapy. The present review offers an overview of recent progress and challenges associated with the intratumoral microbiome and engineered exosomes in the context of cancer research. These discussions are used to inform the construction of a novel framework for engineered exosome-mediated targeted drug delivery, taking advantage of intratumoral microbiota diversity as a strategic asset and thereby providing new opportunities to more effectively treat and manage cancer in the clinic.
Collapse
Affiliation(s)
- Jie Qiu
- Department of Breast and Thyroid Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, 312000, China
| | - Yuancong Jiang
- Department of Breast and Thyroid Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, 312000, China
| | - Nanwei Ye
- Department of Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, 312000, China
| | - Gan Jin
- Department of Vascular Hernia Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, 312000, China
| | - Hao Shi
- Department of Radiotherapy, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, 312000, China
| | - Da Qian
- Department of Burn and Plastic Surgery-Hand Surgery, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, Jiangsu Province, 215500, China
- Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People ' s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang Province, 310014, China
| |
Collapse
|
24
|
Kim HR, Park SJ, Cho YS, Moyo MK, Choi JU, Lee NK, Chung SW, Kweon S, Park J, Kim B, Ko YG, Yeo JH, Lee J, Kim SY, Byun Y. Stimulating macropinocytosis of peptide-drug conjugates through DNA-dependent protein kinase inhibition for treating KRAS-mutant cancer. J Control Release 2024; 372:176-193. [PMID: 38880331 DOI: 10.1016/j.jconrel.2024.06.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
KRAS-mutant cancers, due to their protein targeting complexity, present significant therapeutic hurdles. The identification of the macropinocytic phenotype in these cancers has emerged as a promising alternative therapeutic target. Our study introduces MPD1, an macropinocytosis-targeting peptide-drug conjugates (PDC), which is developed to treat KRAS mutant cancers. This PDC is specifically designed to trigger a positive feedback loop through its caspase-3 cleavable characteristic. However, we observe that this loop is hindered by DNA-PK mediated DNA damage repair processes in cancer cells. To counter this impediment, we employ AZD7648, a DNA-PK inhibitor. Interestingly, the combined treatment of MPD1 and AZD7648 resulted in a 100% complete response rate in KRAS-mutant xenograft model. We focus on the synergic mechanism of it. We discover that AZD7648 specifically enhances macropinocytosis in KRAS-mutant cancer cells. Further analysis uncovers a significant correlation between the increase in macropinocytosis and PI3K signaling, driven by AMPK pathways. Also, AZD7648 reinforces the positive feedback loop, leading to escalated apoptosis and enhanced payload accumulation within tumors. AZD7648 possesses broad applications in augmenting nano-sized drug delivery and preventing DNA repair resistance. The promising efficacy and evident synergy underscore the potential of combining MPD1 with AZD7648 as a strategy for treating KRAS-mutant cancers.
Collapse
Affiliation(s)
- Ha Rin Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.; School of Medicine, Stanford University, CA 94305, United States
| | - Seong Jin Park
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Seok Cho
- College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, United States
| | | | - Jeong Uk Choi
- College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Na Kyeong Lee
- Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Seung Woo Chung
- Depertment of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, United States
| | - Seho Kweon
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jooho Park
- Department of Applied Life Science, Konkuk University, Chungju, 27478, Republic of Korea
| | - Byoungmo Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yoon Gun Ko
- Pharosgen Co.Ltd, Seoul, 05852, Republic of Korea
| | - Joo Hye Yeo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea
| | - Jinu Lee
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, Republic of Korea
| | - Sang Yoon Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Youngro Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea..
| |
Collapse
|
25
|
Quintana J, Kang M, Hu H, Ng TSC, Wojtkiewicz GR, Scott E, Parangi S, Schuemann J, Weissleder R, Miller MA. Extended Pharmacokinetics Improve Site-Specific Prodrug Activation Using Radiation. ACS CENTRAL SCIENCE 2024; 10:1371-1382. [PMID: 39071065 PMCID: PMC11273447 DOI: 10.1021/acscentsci.4c00354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 07/30/2024]
Abstract
Radiotherapy is commonly used to treat cancer, and localized energy deposited by radiotherapy has the potential to chemically uncage prodrugs; however, it has been challenging to demonstrate prodrug activation that is both sustained in vivo and truly localized to tumors without affecting off-target tissues. To address this, we developed a series of novel phenyl-azide-caged, radiation-activated chemotherapy drug-conjugates alongside a computational framework for understanding corresponding pharmacokinetic and pharmacodynamic (PK/PD) behaviors. We especially focused on an albumin-bound prodrug of monomethyl auristatin E (MMAE) and found it blocked tumor growth in mice, delivered a 130-fold greater amount of activated drug to irradiated tumor versus unirradiated tissue, was 7.5-fold more efficient than a non albumin-bound prodrug, and showed no appreciable toxicity compared to free or cathepsin-activatable drugs. These data guided computational modeling of drug action, which indicated that extended pharmacokinetics can improve localized and cumulative drug activation, especially for payloads with low vascular permeability and diffusivity and particularly in patients receiving daily treatments of conventional radiotherapy for weeks. This work thus offers a quantitative PK/PD framework and proof-of-principle experimental demonstration of how extending prodrug circulation can improve its localized activity in vivo.
Collapse
Affiliation(s)
- Jeremy
M. Quintana
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Mikyung Kang
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Huiyu Hu
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Surgery, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Thomas S. C. Ng
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Gregory R. Wojtkiewicz
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
| | - Ella Scott
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
| | - Sareh Parangi
- Department
of Surgery, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Jan Schuemann
- Department
of Radiation Oncology, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Ralph Weissleder
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Miles A. Miller
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
26
|
Ai Y, Tian Y, Qiao J, Wang C, Li H. " Yin-Yang philosophy" for the design of anticancer drug delivery nanoparticles. BIOMATERIALS TRANSLATIONAL 2024; 5:144-156. [PMID: 39351167 PMCID: PMC11438609 DOI: 10.12336/biomatertransl.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/30/2024] [Accepted: 05/20/2024] [Indexed: 10/04/2024]
Abstract
Understanding the in vivo transport process provides guidelines for designing ideal nanoparticles (NPs) with higher efficacy and fewer off-target effects. Many factors, such as particle size, morphology, surface potential, structural stability, and etc., may influence the delivering process of NPs due to the existence of various physiological barriers within the body. Herein, we summarise the distinct influences of NP physicochemical properties on the four consecutive in vivo transport steps: (1) navigating with bloodstream within blood vessels, (2) transport across vasculature walls into tumour tissues, (3) intratumoural transport through the interstitial space, and (4) cellular uptake & intracellular delivery by cancerous cells. We found that the philosophy behind the current consensus for NP design has certain similarities to the "Yin-Yang" theory in traditional Chinese culture. Almost all physicochemical properties, regardless of big or small sizes, long or short length, positive or negative zeta potentials, are double-edged swords. The balance of potential benefits and side effects, drug selectivity and accessibility should be fully considered when optimising particle design, similar to the "Yin-Yang harmony". This paper presents a comprehensive review of the advancements in NPs research, focusing on their distinct features in tumour targeting, drug delivery, and cell uptake. Additionally, it deliberates on future developmental trends and potential obstacles, thereby aiming to uncover the ways these characteristics influence the NPs' biological activity and provide theoretical guidance for the targeted delivery of NPs.
Collapse
Affiliation(s)
- Yanwen Ai
- School of Lifesciences, Shanghai University, Shanghai, China
| | - Yuan Tian
- School of Lifesciences, Shanghai University, Shanghai, China
| | - Jiaming Qiao
- School of Lifesciences, Shanghai University, Shanghai, China
| | - Changnan Wang
- School of Lifesciences, Shanghai University, Shanghai, China
| | - Huafei Li
- School of Lifesciences, Shanghai University, Shanghai, China
| |
Collapse
|
27
|
Wu Y, Hu X, Wei Z, Lin Q. Cellular Regulation of Macropinocytosis. Int J Mol Sci 2024; 25:6963. [PMID: 39000072 PMCID: PMC11241348 DOI: 10.3390/ijms25136963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/17/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024] Open
Abstract
Interest in macropinocytosis has risen in recent years owing to its function in tumorigenesis, immune reaction, and viral infection. Cancer cells utilize macropinocytosis to acquire nutrients to support their uncontrolled proliferation and energy consumption. Macropinocytosis, a highly dynamic endocytic and vesicular process, is regulated by a series of cellular signaling pathways. The activation of small GTPases in conjunction with phosphoinositide signaling pivotally regulates the process of macropinocytosis. In this review, we summarize important findings about the regulation of macropinocytosis and provide information to increase our understanding of the regulatory mechanism underlying it.
Collapse
Affiliation(s)
| | | | | | - Qiong Lin
- School of Medicine, Jiangsu University, Zhenjiang 212013, China; (Y.W.); (X.H.); (Z.W.)
| |
Collapse
|
28
|
Xu Z, Lin H, Dai J, Wen X, Yu X, Xu C, Ruan G. Protein-nanoparticle co-assembly supraparticles for drug delivery: Ultrahigh drug loading and colloidal stability, and instant and complete lysosomal drug release. Int J Pharm 2024; 658:124231. [PMID: 38759741 DOI: 10.1016/j.ijpharm.2024.124231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/06/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Two frequent problems hindering clinical translation of nanomedicine are low drug loading and low colloidal stability. Previous efforts to achieve ultrahigh drug loading (>30 %) introduce new hurdles, including lower colloidal stability and others, for clinical translation. Herein, we report a new class of drug nano-carriers based on our recent finding in protein-nanoparticle co-assembly supraparticle (PNCAS), with both ultrahigh drug loading (58 % for doxorubicin, i.e., DOX) and ultrahigh colloidal stability (no significant change in hydrodynamic size after one year). We further show that our PNCAS-based drug nano-carrier possesses a built-in environment-responsive drug release feature: once in lysosomes, the loaded drug molecules are released instantly (<1 min) and completely (∼100 %). Our PNCAS-based drug delivery system is spontaneously formed by simple mixing of hydrophobic nanoparticles, albumin and drugs. Several issues related to industrial production are studied. The ultrahigh drug loading and stability of DOX-loaded PNCAS enabled the delivery of an exceptionally high dose of DOX into a mouse model of breast cancer, yielding high efficacy and no observed toxicity. With further developments, our PNCAS-based delivery systems could serve as a platform technology to meet the multiple requirements of clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Zixing Xu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210093, China; Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China; Xi'an Jiaotong-Liverpool University & University of Liverpool Joint Center of Pharmacology and Therapeutics, Suzhou 215123, China
| | - Huoyue Lin
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210093, China
| | - Jie Dai
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210093, China
| | - Xiaowei Wen
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China; Xi'an Jiaotong-Liverpool University & University of Liverpool Joint Center of Pharmacology and Therapeutics, Suzhou 215123, China; Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xiaoya Yu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210093, China
| | - Can Xu
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Gang Ruan
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210093, China; Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China; Xi'an Jiaotong-Liverpool University & University of Liverpool Joint Center of Pharmacology and Therapeutics, Suzhou 215123, China; Institute of Materials Engineering of Nanjing University, Nantong 210033, China.
| |
Collapse
|
29
|
Chen Y, Xiang A, Ma Y, Qian Z, Ma Z, Zhou L, Wang J, Li H, Zhou M, Hou Z, Liu X, Zhang C, Tang L, Ju S, Fan W. Photoactivated Formation of an Extravascular Dynamic Hydrogel as an Intelligent Blood Flow Regulator to Reprogram the Immunogenic Landscape. NANO LETTERS 2024; 24:5690-5698. [PMID: 38700237 DOI: 10.1021/acs.nanolett.4c00376] [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: 05/05/2024]
Abstract
Long-term tumor starvation may be a potential strategy to elevate the antitumor immune response by depriving nutrients. However, combining long-term starvation therapy with immunotherapy often yields limited efficacy due to the blockage of immune cell migration pathways. Herein, an intelligent blood flow regulator (BFR) is first established through photoactivated in situ formation of the extravascular dynamic hydrogel to compress blood vessels, which can induce long-term tumor starvation to elicit metabolic stress in tumor cells without affecting immune cell migration pathways. By leveraging methacrylate-modified nanophotosensitizers (HMMAN) and biodegradable gelatin methacrylate (GelMA), the developed extravascular hydrogel dynamically regulates blood flow via enzymatic degradation. Additionally, aPD-L1 loaded into HMMAN continuously blocks immune checkpoints. Systematic in vivo experiments demonstrate that the combination of immune checkpoint blockade (ICB) and BFR-induced metabolic stress (BIMS) significantly delays the progression of Lewis lung and breast cancers by reshaping the tumor immunogenic landscape and enhancing antitumor immune responses.
Collapse
Affiliation(s)
- Yue Chen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Ajun Xiang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Yuanyuan Ma
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhengzheng Qian
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Zerui Ma
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Liming Zhou
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Jue Wang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hao Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Mei Zhou
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhenyu Hou
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xin Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Can Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | | | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
30
|
Wilson J, Kimmel B, Arora K, Chada N, Bharti V, Kwiatkowski A, Finklestein J, Hanna A, Arner E, Sheehy T, Pastora L, Yang J, Pagendarm H, Stone P, Taylor B, Hubert L, Gibson-Corley K, May J, McLean J, Rathmell J, Richmond A, Rathmell W, Balko J, Fingleton B, Hargrove-Wiley E. Programable Albumin-Hitchhiking Nanobodies Enhance the Delivery of STING Agonists to Potentiate Cancer Immunotherapy. RESEARCH SQUARE 2024:rs.3.rs-3243545. [PMID: 38766114 PMCID: PMC11100900 DOI: 10.21203/rs.3.rs-3243545/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Stimulator of interferon genes (STING) is a promising target for potentiating antitumor immunity, but multiple pharmacological barriers limit the clinical utility, efficacy, and/or safety of STING agonists. Here we describe a modular platform for systemic administration of STING agonists based on nanobodies engineered for in situ hitchhiking of agonist cargo on serum albumin. Using site-selective bioconjugation chemistries to produce molecularly defined products, we found that covalent conjugation of a STING agonist to anti-albumin nanobodies improved pharmacokinetics and increased cargo accumulation in tumor tissue, stimulating innate immune programs that increased the infiltration of activated natural killer cells and T cells, which potently inhibited tumor growth in multiple mouse tumor models. We also demonstrated the programmability of the platform through the recombinant integration of a second nanobody domain that targeted programmed cell death ligand-1 (PD-L1), which further increased cargo delivery to tumor sites while also blocking immunosuppressive PD-1/PD-L1 interactions. This bivalent nanobody carrier for covalently conjugated STING agonists stimulated robust antigen-specific T cell responses and long-lasting immunological memory, conferred enhanced therapeutic efficacy, and was effective as a neoadjuvant treatment for improving responses to adoptive T cell transfer therapy. Albumin-hitchhiking nanobodies thus offer an enabling, multimodal, and programmable platform for systemic delivery of STING agonists with potential to augment responses to multiple immunotherapeutic modalities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ann Hanna
- Vanderbilt University Medical Center
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Li X, Zhong H, Zheng S, Mu J, Yu N, Guo S. Tumor-penetrating iRGD facilitates penetration of poly(floxuridine-ketal)-based nanomedicine for enhanced pancreatic cancer therapy. J Control Release 2024; 369:444-457. [PMID: 38575076 DOI: 10.1016/j.jconrel.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/12/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Efficient intratumoral penetration is essential for nanomedicine to eradicate pancreatic tumors. Although nanomedicine can enter the perivascular space of pancreatic tumors, their access to distal tumor cells, aloof from the vessels, remains a formidable challenge. Here, we synthesized an acid-activatable macromolecular prodrug of floxuridine (FUDR)-poly(FUDR-ketal), engineered a micellar nanomedicine of FUDR, and intravenously co-administered the nanomedicine with the tumor-penetrating peptide iRGD for enhanced treatment of pancreatic tumor. A FUDR-derived mono-isopropenyl ether was synthesized and underwent self-addition polymerization to afford the hydrophobic poly(FUDR-ketal), which was subsequently co-assembled with amphiphilic DSPE-mPEG into the micellar nanomedicine with size of 12 nm and drug content of 56.8 wt% using nanoprecipitation technique. The acetone-based ketal-linked poly(FUDR-ketal) was triggered by acid to release FUDR to inhibit cell proliferation. In an orthotopic pancreatic tumor model derived from KPC (KrasLSL-G12D/+; Trp53LSL-R172H/+; Pdx1-Cre) cells that overexpress neuropilin-1 (NRP-1) receptor, iRGD improved penetration of FUDR nanomedicine into tumor parenchyma and potentiated the therapeutic efficacy. Our nanoplatform, along with iRGD, thus appears to be promising for efficient penetration and activation of acid-responsive nanomedicines for enhanced pancreatic cancer therapy.
Collapse
Affiliation(s)
- Xingwei Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Haiping Zhong
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Shujing Zheng
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jingqing Mu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China; Tianjin Aier Eye Hospital, Tianjin 300190, China
| | - Na Yu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China; Jingjinji National Center of Technology Innovation, Beijing 100094, China.
| | - Shutao Guo
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China.
| |
Collapse
|
32
|
Liang G, Cao W, Tang D, Zhang H, Yu Y, Ding J, Karges J, Xiao H. Nanomedomics. ACS NANO 2024; 18:10979-11024. [PMID: 38635910 DOI: 10.1021/acsnano.3c11154] [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: 04/20/2024]
Abstract
Nanomaterials have attractive physicochemical properties. A variety of nanomaterials such as inorganic, lipid, polymers, and protein nanoparticles have been widely developed for nanomedicine via chemical conjugation or physical encapsulation of bioactive molecules. Superior to traditional drugs, nanomedicines offer high biocompatibility, good water solubility, long blood circulation times, and tumor-targeting properties. Capitalizing on this, several nanoformulations have already been clinically approved and many others are currently being studied in clinical trials. Despite their undoubtful success, the molecular mechanism of action of the vast majority of nanomedicines remains poorly understood. To tackle this limitation, herein, this review critically discusses the strategy of applying multiomics analysis to study the mechanism of action of nanomedicines, named nanomedomics, including advantages, applications, and future directions. A comprehensive understanding of the molecular mechanism could provide valuable insight and therefore foster the development and clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Ganghao Liang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Wanqing Cao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hanchen Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
33
|
Taylor KS, McMonagle MM, Guy SC, Human-McKinnon AM, Asamizu S, Fletcher HJ, Davis BW, Suyama TL. Albumin-ruthenium catalyst conjugate for bio-orthogonal uncaging of alloc group. Org Biomol Chem 2024; 22:2992-3000. [PMID: 38526322 DOI: 10.1039/d4ob00234b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
The employment of antibodies as a targeted drug delivery vehicle has proven successful which is exemplified by the emergence of antibody-drug conjugates (ADCs). However, ADCs are not without their shortcomings. Improvements may be made to the ADC platform by decoupling the cytotoxic drug from the delivery vehicle and conjugating an organometallic catalyst in its place. The resulting protein-metal catalyst conjugate was designed to uncage the masked cytotoxin administered as a separate entity. Macropinocytosis of albumin by cancerous cells suggests the potential of albumin acting as the tumor-targeting delivery vehicle. Herein reported are the first preparation and demonstration of ruthenium catalysts with cyclopentadienyl and quinoline-based ligands conjugated to albumin. The effective uncaging abilities were demonstrated on allyloxy carbamate (alloc)-protected rhodamine 110 and doxorubicin, providing a promising catalytic scaffold for the advancement of selective drug delivery methods in the future.
Collapse
Affiliation(s)
- Kimberly S Taylor
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Madison M McMonagle
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Schaelee C Guy
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Ariana M Human-McKinnon
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Shumpei Asamizu
- Engineering Biology Research Center, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Heidi J Fletcher
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Bradley W Davis
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Takashi L Suyama
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| |
Collapse
|
34
|
Guo X, Lou W, Xu Y, Zhuang R, Yao L, Wu J, Fu D, Zhang J, Liu J, Rong Y, Jin D, Wu W, Xu X, Ji Y, Wu L, Lv M, Yao X, Liu X, Wang D, Kuang T, Liu L, Wang W, Liu T, Zhou Y. Efficacy of nab‑paclitaxel vs. Gemcitabine in combination with S‑1 for advanced pancreatic cancer: A multicenter phase II randomized trial. Oncol Lett 2024; 27:161. [PMID: 38449794 PMCID: PMC10915801 DOI: 10.3892/ol.2024.14293] [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: 09/25/2023] [Accepted: 01/17/2024] [Indexed: 03/08/2024] Open
Abstract
Patients with advanced pancreatic cancer (PC) need a cost-effective treatment regimen. The present study was designed to compare the efficacy and safety of nab-paclitaxel plus S-1 (AS) and gemcitabine plus S-1 (GS) regimens in patients with chemotherapy-naïve advanced PC. In this open-label, multicenter, randomized study named AvGmPC, eligible patients with chemotherapy-naïve advanced PC were randomly assigned (1:1) to receive AS (125 mg/m2 nab-paclitaxel, days 1 and 8; 80-120 mg S-1, days 1-14) or GS (1,000 mg/m2 gemcitabine, days 1 and 8; 80-120 mg S-1, days 1-14). The treatment was administered every 3 weeks until intolerable toxicity or disease progression occurred. The primary endpoint was progression-free survival (PFS). Between December 2018 and March 2022, 101 of 106 randomized patients were treated and evaluated for analysis (AS, n=49; GS, n=52). As of the data cutoff, the median follow-up time was 11.37 months [95% confidence interval (CI), 9.31-13.24]. The median PFS was 7.16 months (95% CI, 5.19-12.32) for patients treated with AS and 6.41 months (95% CI, 3.72-8.84) for patients treated with GS (HR=0.78; 95% CI, 0.51-1.21; P=0.264). The AS regimen showed a slightly improved overall survival (OS; 13.27 vs. 10.64 months) and a significantly improved ORR (44.90 vs. 15.38%; P=0.001) compared with the GS regimen. In the subgroup analyses, PFS and OS benefits were observed in patients treated with the AS regimen who had KRAS gene mutations and high C-reactive protein (CRP) levels (≥5 mg/l). The most common grade ≥3 adverse events were neutropenia, anemia and alopecia in the two groups. Thrombocytopenia occurred more frequently in the GS group than in the AS group. While the study did not meet the primary endpoint, the response benefit observed for AS may be suggestive of meaningful clinical activity in this population. In particular, promising survival benefits were observed in the subsets of patients with KRAS gene mutations and high CRP levels, which is encouraging and warrants further investigation. This trial was retrospectively registered as ChiCTR1900024588 on July 18, 2019.
Collapse
Affiliation(s)
- Xi Guo
- Department of Medical Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
- Cancer Center, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
- Center of Evidence-Based Medicine, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Wenhui Lou
- Department of General Surgery, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Yaolin Xu
- Department of General Surgery, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Rongyuan Zhuang
- Department of Medical Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
- Cancer Center, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
- Center of Evidence-Based Medicine, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Lie Yao
- Department of Pancreatic Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Junwei Wu
- Oncology Department, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Deliang Fu
- Department of Pancreatic Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Jun Zhang
- Oncology Department, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Jing Liu
- Oncology Department, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yefei Rong
- Department of General Surgery, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Dayong Jin
- Department of General Surgery, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Wenchuan Wu
- Department of General Surgery, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Xuefeng Xu
- Department of General Surgery, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Lili Wu
- Department of Radiotherapy, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Minzhi Lv
- Department of Biostatistics, Clinical Research Unit, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Xiuzhong Yao
- Department of Radiology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Xiaowei Liu
- Department of Anti-tumor Business, Shi Yao Group European Pharmaceutical Co., Ltd., Shijiazhuang, Hebei 050035, P.R. China
| | - Dansong Wang
- Department of General Surgery, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Tiantao Kuang
- Department of General Surgery, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Liang Liu
- Department of General Surgery, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Wenquan Wang
- Department of General Surgery, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
- Cancer Center, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
- Center of Evidence-Based Medicine, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| | - Yuhong Zhou
- Department of Medical Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
- Cancer Center, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
35
|
Chen Y, Ma Y, Shi K, Chen H, Han X, Wei C, Lyu Y, Huang Y, Yu R, Song Y, Song Q, Jiang J, Feng J, Lin Y, Chen J, Chen H, Zheng G, Gao X, Jiang G. Self-Disassembling and Oxygen-Generating Porphyrin-Lipoprotein Nanoparticle for Targeted Glioblastoma Resection and Enhanced Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307454. [PMID: 38299428 DOI: 10.1002/adma.202307454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/15/2023] [Indexed: 02/02/2024]
Abstract
The dismal prognosis for glioblastoma multiform (GBM) patients is primarily attributed to the highly invasive tumor residual that remained after surgical intervention. The development of precise intraoperative imaging and postoperative residual removal techniques will facilitate the gross total elimination of GBM. Here, a self-disassembling porphyrin lipoprotein-coated calcium peroxide nanoparticles (PLCNP) is developed to target GBM via macropinocytosis, allowing for fluorescence-guided surgery of GBM and improving photodynamic treatment (PDT) of GBM residual by alleviating hypoxia. By reducing self-quenching and enhancing lysosome escape efficiency, the incorporation of calcium peroxide (CaO2) cores in PLCNP amplifies the fluorescence intensity of porphyrin-lipid. Furthermore, the CaO2 core has diminished tumor hypoxia and improves the PDT efficacy of PLCNP, enabling low-dose PDT and reversing tumor progression induced by hypoxia aggravation following PDT. Taken together, this self-disassembling and oxygen-generating porphyrin-lipoprotein nanoparticle may serve as a promising all-in-one nanotheranostic platform for guiding precise GBM excision and empowering post-operative PDT, providing a clinically applicable strategy to combat GBM in a safe and effective manner.
Collapse
Affiliation(s)
- Yaoxing Chen
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yuxiao Ma
- Brain Injury Centre, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Kexin Shi
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Huan Chen
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Xiao Han
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Chenxuan Wei
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yingqi Lyu
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yukun Huang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Renhe Yu
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yun Song
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Qingxiang Song
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Jiyao Jiang
- Brain Injury Centre, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Junfeng Feng
- Brain Injury Centre, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Yingying Lin
- Brain Injury Centre, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Jun Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
- Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201210, China
| | - Gang Zheng
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| |
Collapse
|
36
|
Xie Z, Ye J, Gao X, Chen H, Chen M, Lian J, Ma J, Wang H. Evaluation of nanoparticle albumin-bound paclitaxel loaded macrophages for glioblastoma treatment based on a microfluidic chip. Front Bioeng Biotechnol 2024; 12:1361682. [PMID: 38562665 PMCID: PMC10982336 DOI: 10.3389/fbioe.2024.1361682] [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: 12/26/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction: Glioblastoma (GBM) is a primary brain malignancy with a dismal prognosis and remains incurable at present. In this study, macrophages (MΦ) were developed to carry nanoparticle albumin-bound paclitaxel (nab-PTX) to form nab-PTX/MΦ. The aim of this study is to use a GBM-on-a-chip to evaluate the anti-GBM effects of nab-PTX/MΦ. Methods: In this study, we constructed nab-PTX/MΦ by incubating live MΦ with nab-PTX. We developed a microfluidic chip to co-culture GBM cells and human umbilical vein endothelial cells, mimicking the simplified blood-brain barrier and GBM. Using a syringe pump, we perform sustainable perfusion of nutrient media. To evaluate the anti-GBM effects nab-PTX/MΦ, we treated the GBM-on-a-chip model with nab-PTX/MΦ and investigated GBM cell proliferation, migration, and spheroid formation. Results: At the chosen concentration, nab-PTX did not significantly affect the viability, chemotaxis and migration of MΦ. The uptake of nab-PTX by MΦ occurred within 1 h of incubation and almost reached saturation at 6 h. Additionally, nab-PTX/MΦ exhibited the M1 phenotype, which inhibits tumor progression. Following phagocytosis, MΦ were able to release nab-PTX, and the release of nab-PTX by MΦ had nearly reached its limit at 48 h. Compared with control group and blank MΦ group, individual nab-PTX group and nab-PTX/MΦ group could inhibit tumor proliferation, invasion and spheroid formation. Meanwhile, the anti-GBM effect of nab-PTX/MΦ was more significant than nab-PTX. Discussion: Our findings demonstrate that nab-PTX/MΦ has a significant anti-GBM effect compared to individual nab-PTX or MΦ administration, suggesting MΦ as potential drug delivery vectors for GBM therapy. Furthermore, the developed GBM-on-a-chip model provides a potential ex vivo platform for innovative cell-based therapies and tailored therapeutic strategies for GBM.
Collapse
Affiliation(s)
- Zuorun Xie
- Department of Neurosurgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Junyi Ye
- Department of Neurosurgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xinghua Gao
- Materials Genome Institute, Shanghai University, Shanghai, China
| | - Hang Chen
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Maosong Chen
- Department of Neurosurgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jiangfang Lian
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jingyun Ma
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Hongcai Wang
- Department of Neurosurgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
37
|
Liao YN, Gai YZ, Qian LH, Pan H, Zhang YF, Li P, Guo Y, Li SX, Nie HZ. Progesterone receptor potentiates macropinocytosis through CDC42 in pancreatic ductal adenocarcinoma. Oncogenesis 2024; 13:10. [PMID: 38424455 PMCID: PMC10904380 DOI: 10.1038/s41389-024-00512-7] [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: 10/18/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
Endocrine receptors play an essential role in tumor metabolic reprogramming and represent a promising therapeutic avenue in pancreatic ductal adenocarcinoma (PDAC). PDAC is characterized by a nutrient-deprived microenvironment. To meet their ascendant energy demands, cancer cells can internalize extracellular proteins via macropinocytosis. However, the roles of endocrine receptors in macropinocytosis are not clear. In this study, we found that progesterone receptor (PGR), a steroid-responsive nuclear receptor, is highly expressed in PDAC tissues obtained from both patients and transgenic LSL-KrasG12D/+; LSL-Trp53R172H/+; PDX1-cre (KPC) mice. Moreover, PGR knockdown restrained PDAC cell survival and tumor growth both in vitro and in vivo. Genetic and pharmacological PGR inhibition resulted in a marked attenuation of macropinocytosis in PDAC cells and subcutaneous tumor models, indicating the involvement of this receptor in macropinocytosis regulation. Mechanistically, PGR upregulated CDC42, a critical regulator in macropinocytosis, through PGR-mediated transcriptional activation. These data deepen the understanding of how the endocrine system influences tumor progression via a non-classical pathway and provide a novel therapeutic option for patients with PDAC.
Collapse
Affiliation(s)
- Ying-Na Liao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Yan-Zhi Gai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Li-Heng Qian
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Hong Pan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Yi-Fan Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Pin Li
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 20030, P.R. China
| | - Ying Guo
- Radiology Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China.
| | - Shu-Xin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China.
| | - Hui-Zhen Nie
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China.
| |
Collapse
|
38
|
Chen K, Cheng X, Xue S, Chen J, Zhang X, Qi Y, Chen R, Zhang Y, Wang H, Li W, Cheng G, Huang Y, Xiong Y, Chen L, Mu C, Gu M. Albumin conjugation promotes arsenic trioxide transport through alkaline phosphatase-associated transcytosis in MUC4 wildtype pancreatic cancer cells. Int J Biol Macromol 2024; 257:128756. [PMID: 38092098 DOI: 10.1016/j.ijbiomac.2023.128756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023]
Abstract
Pancreatic cancer (PC) has a poor prognosis due to chemotherapy resistance and unfavorable drug transportation. Albumin conjugates are commonly used as drug carriers to overcome these obstacles. However, membrane-bound glycoprotein mucin 4 (MUC4) has emerged as a promising biomarker among the genetic mutations affecting albumin conjugates therapeutic window. Human serum albumin-conjugated arsenic trioxide (HSA-ATO) has shown potential in treating solid tumors but is limited in PC therapy due to unclear targets and mechanisms. This study investigated the transport mechanisms and therapeutic efficacy of HSA-ATO in PC cells with different MUC4 mutation statuses. Results revealed improved penetration of ATO into PC tumors through conjugated with HSA. However, MUC4 mutation significantly affected treatment sensitivity and HSA-ATO uptake both in vitro and in vivo. Mutant MUC4 cells exhibited over ten times higher IC50 for HSA-ATO and approximately half the uptake compared to wildtype cells. Further research demonstrated that ALPL activation by HSA-ATO enhanced transcytosis in wildtype MUC4 PC cells but not in mutant MUC4 cells, leading to impaired uptake and weaker antitumor effects. Reprogramming the transport process holds potential for enhancing albumin conjugate efficacy in PC patients with different MUC4 mutation statuses, paving the way for stratified treatment using these delivery vehicles.
Collapse
Affiliation(s)
- Kaidi Chen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Xiao Cheng
- Huzhou Institute for Food and Drug Control, Huzhou 313000, PR China
| | - Shuai Xue
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Junyan Chen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Xu Zhang
- Zhejiang Heze Pharmaceutical Technology Co., Ltd., Hangzhou 310018, Zhejiang, PR China
| | - Yuwei Qi
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Rong Chen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Yan Zhang
- Department of Pharmacy, Hangzhou Red Cross Hospital, Hangzhou 310003, Zhejiang, PR China
| | - Hangjie Wang
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Wei Li
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Guilin Cheng
- Department of Pharmacy, Hangzhou Red Cross Hospital, Hangzhou 310003, Zhejiang, PR China
| | - Ye Huang
- Department of Pharmacy, Zhejiang Provincial Dermatology Hospital, Huzhou 313200, Zhejiang, PR China
| | - Yang Xiong
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China; Department of Pharmacy, Hangzhou Red Cross Hospital, Hangzhou 310003, Zhejiang, PR China
| | - Liping Chen
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, PR China; School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, Jiangsu, PR China.
| | - Chaofeng Mu
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China.
| | - Mancang Gu
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China.
| |
Collapse
|
39
|
Chan WJ, Li H. Recent advances in nano/micro systems for improved circulation stability, enhanced tumor targeting, penetration, and intracellular drug delivery: a review. Biomed Phys Eng Express 2024; 10:022001. [PMID: 38086099 DOI: 10.1088/2057-1976/ad14f0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 12/12/2023] [Indexed: 01/17/2024]
Abstract
In recent years, nanoparticles (NPs) have been extensively developed as drug carriers to overcome the limitations of cancer therapeutics. However, there are several biological barriers to nanomedicines, which include the lack of stability in circulation, limited target specificity, low penetration into tumors and insufficient cellular uptake, restricting the active targeting toward tumors of nanomedicines. To address these challenges, a variety of promising strategies were developed recently, as they can be designed to improve NP accumulation and penetration in tumor tissues, circulation stability, tumor targeting, and intracellular uptake. In this Review, we summarized nanomaterials developed in recent three years that could be utilized to improve drug delivery for cancer treatments.
Collapse
Affiliation(s)
- Wei-Jen Chan
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States of America
| | - Huatian Li
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States of America
| |
Collapse
|
40
|
Qin M, Xia H, Xu W, Chen B, Wang Y. The spatiotemporal journey of nanomedicines in solid tumors on their therapeutic efficacy. Adv Drug Deliv Rev 2023; 203:115137. [PMID: 37949414 DOI: 10.1016/j.addr.2023.115137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
The rapid development of nanomedicines is revolutionizing the landscape of cancer treatment, while effectively delivering them into solid tumors remains a formidable challenge. Currently, there is a huge disconnect on therapeutic response between regulatory approved nanomedicines and laboratory reported nanoparticles. The discrepancy is mainly resulted from the failure of using the classic overall pharmacokinetics behaviors of nanomedicines in tumors to predict the antitumor efficacy. Increasing evidence has revealed that the therapeutic efficacy predominantly relies on the intratumoral spatiotemporal distribution of nanomedicines. This review focuses on the spatiotemporal distribution of systemically administered chemotherapeutic nanomedicines in solid tumor. Firstly, the intratumoral biological barriers that regulate the spatiotemporal distribution of nanomedicines are described in detail. Next, the influences on antitumor efficacy caused by the spatial distribution and temporal drug release of nanomedicines are emphatically analyzed. Then, current methodologies for evaluating the spatiotemporal distribution of nanomedicines are summarized. Finally, the advanced strategies to positively modulate the spatiotemporal distribution of nanomedicines for an optimal tumor therapy are comprehensively reviewed.
Collapse
Affiliation(s)
- Mengmeng Qin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Heming Xia
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wenhao Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China; Chemical Biology Center, Peking University, Beijing, China.
| |
Collapse
|
41
|
Lee B, Park OK, Pan L, Kim K, Kang T, Kim H, Lee N, Choi SH, Hyeon T. Co-Delivery of Metabolic Modulators Leads to Simultaneous Lactate Metabolism Inhibition and Intracellular Acidification for Synergistic Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2305512. [PMID: 37487702 DOI: 10.1002/adma.202305512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/13/2023] [Indexed: 07/26/2023]
Abstract
Simultaneous lactate metabolism inhibition and intracellular acidification (LIIA) is a promising approach for inducing tumor regression by depleting ATP. However, given the limited efficacy of individual metabolic modulators, a combination of various modulators is required for highly efficient LIIA. Herein, a co-delivery system that combines lactate transporter inhibitor, glucose oxidase, and O2 -evolving nanoparticles is proposed. As a vehicle, a facile room-temperature synthetic method for large-pore mesoporous silica nanoparticles (L-MSNs) is developed. O2 -evolving nanoparticles are then conjugated onto L-MSNs, followed by immobilizing the lactate transporter inhibitor and glucose oxidase inside the pores of L-MSNs. To load the lactate transporter inhibitor, which is too small to be directly loaded into the large pores, it is encapsulated in albumin by controlling the albumin conformation before being loaded into L-MSNs. Notably, inhibiting lactate efflux shifts the glucose consumption mechanism from lactate metabolism to glucose oxidase reaction, which eliminates glucose and produces acid. This leads to synergistic LIIA and subsequent ATP depletion in cancer cells. Consequently, L-MSN-based co-delivery of modulators for LIIA shows high anticancer efficacy in several mouse tumor models without toxicity in normal tissues. This study provides new insights into co-delivery of small-molecule drugs, proteins, and nanoparticles for synergistic metabolic modulation in tumors.
Collapse
Affiliation(s)
- Bowon Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ok Kyu Park
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Limin Pan
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kang Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Taegyu Kang
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyunjoong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nohyun Lee
- School of Advanced Materials Engineering, Kookmin University, Seoul, 02707, Republic of Korea
| | - Seung Hong Choi
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
42
|
Shi Z, Luo M, Huang Q, Ding C, Wang W, Wu Y, Luo J, Lin C, Chen T, Zeng X, Mei L, Zhao Y, Chen H. NIR-dye bridged human serum albumin reassemblies for effective photothermal therapy of tumor. Nat Commun 2023; 14:6567. [PMID: 37848496 PMCID: PMC10582160 DOI: 10.1038/s41467-023-42399-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 10/10/2023] [Indexed: 10/19/2023] Open
Abstract
Human serum albumin (HSA) based drug delivery platforms that feature desirable biocompatibility and pharmacokinetic property are rapidly developed for tumor-targeted drug delivery. Even though various HSA-based platforms have been established, it is still of great significance to develop more efficient preparation technology to broaden the therapeutic applications of HSA-based nano-carriers. Here we report a bridging strategy that unfastens HSA to polypeptide chains and subsequently crosslinks these chains by a bridge-like molecule (BPY-Mal2) to afford the HSA reassemblies formulation (BPY@HSA) with enhanced loading capacity, endowing the BPY@HSA with uniformed size, high photothermal efficacy, and favorable therapeutic features. Both in vitro and in vivo studies demonstrate that the BPY@HSA presents higher delivery efficacy and more prominent photothermal therapeutic performance than that of the conventionally prepared formulation. The feasibility in preparation, stability, high photothermal conversion efficacy, and biocompatibility of BPY@HSA may facilitate it as an efficient photothermal agents (PTAs) for tumor photothermal therapy (PTT). This work provides a facile strategy to enhance the loading capacity of HSA-based crosslinking platforms in order to improve delivery efficacy and therapeutic effect.
Collapse
Affiliation(s)
- Zhaoqing Shi
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Miaomiao Luo
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Qili Huang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Chendi Ding
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Wenyan Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yinglong Wu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Jingjing Luo
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Chuchu Lin
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Ting Chen
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Xiaowei Zeng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China.
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore.
| | - Hongzhong Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore.
| |
Collapse
|
43
|
Ma X, Tang Y, Wang C, Li Y, Zhang J, Luo Y, Xu Z, Wu F, Wang S. Interpretable XGBoost-SHAP Model Predicts Nanoparticles Delivery Efficiency Based on Tumor Genomic Mutations and Nanoparticle Properties. ACS APPLIED BIO MATERIALS 2023; 6:4326-4335. [PMID: 37683105 DOI: 10.1021/acsabm.3c00527] [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: 09/10/2023]
Abstract
Understanding the complex interaction between nanoparticles (NPs) and tumors in vivo and how it dominates the delivery efficiency of NPs is critical for the translation of nanomedicine. Herein, we proposed an interpretable XGBoost-SHAP model by integrating the information on NPs physicochemical properties and tumor genomic profile to predict the delivery efficiency. The correlation coefficients were 0.66, 0.75, and 0.54 for the prediction of maximum delivery efficiency, delivery efficiency at 24 and 168 h postinjection for test sets. The analysis of the feature importance revealed that the tumor genomic mutations and their interaction with NPs properties played important roles in the delivery of NPs. The biological pathways of the NP-delivery-related genes were further explored through gene ontology enrichment analysis. Our work provides a pipeline to predict and explain the delivery efficiency of NPs to heterogeneous tumors and highlights the power of simultaneously using omics data and interpretable machine learning algorithms for discovering interactions between NPs and individual tumors, which is important for the development of personalized precision nanomedicine.
Collapse
Affiliation(s)
- Xingqun Ma
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
- Department of Oncology, Nanjing Baiyi Hospital, Jinling Clinical College of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yuxia Tang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Chuanbing Wang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yang Li
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Jiulou Zhang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yafei Luo
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Ziqing Xu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Feiyun Wu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Shouju Wang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| |
Collapse
|
44
|
Lin YW, Lin TT, Chen CH, Wang RH, Lin YH, Tseng TY, Zhuang YJ, Tang SY, Lin YC, Pang JY, Chakravarthy RD, Lin HC, Tzou SC, Chao JI. Enhancing Efficacy of Albumin-Bound Paclitaxel for Human Lung and Colorectal Cancers through Autophagy Receptor Sequestosome 1 (SQSTM1)/p62-Mediated Nanodrug Delivery and Cancer therapy. ACS NANO 2023; 17:19033-19051. [PMID: 37737568 DOI: 10.1021/acsnano.3c04739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Selective autophagy is a defense mechanism by which foreign pathogens and abnormal substances are processed to maintain cellular homeostasis. Sequestosome 1 (SQSTM1)/p62, a vital selective autophagy receptor, recruits ubiquitinated cargo to form autophagosomes for lysosomal degradation. Nab-PTX is an albumin-bound paclitaxel nanoparticle used in clinical cancer therapy. However, the role of SQSTM1 in regulating the delivery and efficacy of nanodrugs remains unclear. Here we showed that SQSTM1 plays a crucial role in Nab-PTX drug delivery and efficacy in human lung and colorectal cancers. Nab-PTX induces SQSTM1 phosphorylation at Ser403, which facilitates its incorporation into the selective autophagy of nanoparticles, known as nanoparticulophagy. Nab-PTX increased LC3-II protein expression, which triggered autophagosome formation. SQSTM1 enhanced Nab-PTX recognition to form autophagosomes, which were delivered to lysosomes for albumin degradation, thereby releasing PTX to induce mitotic catastrophe and apoptosis. Knockout of SQSTM1 downregulated Nab-PTX-induced mitotic catastrophe, apoptosis, and tumor inhibition in vitro and in vivo and inhibited Nab-PTX-induced caspase 3 activation via a p53-independent pathway. Ectopic expression of SQSTM1 by transfection of an SQSTM1-GFP vector restored the drug efficacy of Nab-PTX. Importantly, SQSTM1 is highly expressed in advanced lung and colorectal tumors and is associated with poor overall survival in clinical patients. Targeting SQSTM1 may provide an important strategy to improve nanodrug efficacy in clinical cancer therapy. This study demonstrates the enhanced efficacy of Nab-PTX for human lung and colorectal cancers via SQSTM1-mediated nanodrug delivery.
Collapse
Affiliation(s)
- Yu-Wei Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Tzu-Ting Lin
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Chien-Hung Chen
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Rou-Hsin Wang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Ya-Hui Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Tzu-Yen Tseng
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Yan-Jun Zhuang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Sheng-Yueh Tang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Yen-Cheng Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Jiun-Yu Pang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Rajan Deepan Chakravarthy
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| | - Hsin-Chieh Lin
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| | - Shey-Cherng Tzou
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Jui-I Chao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Center For Intelligent Drug Systems and Smart Bio-devices, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| |
Collapse
|
45
|
Cui T, Corrales-Guerrero S, Castro-Aceituno V, Nair S, Maneval DC, Monnig C, Kearney P, Ellis S, Raheja N, Raheja N, Williams TM. JNTX-101, a novel albumin-encapsulated gemcitabine prodrug, is efficacious and operates via caveolin-1-mediated endocytosis. Mol Ther Oncolytics 2023; 30:181-192. [PMID: 37674628 PMCID: PMC10477748 DOI: 10.1016/j.omto.2023.08.008] [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: 05/18/2023] [Accepted: 08/15/2023] [Indexed: 09/08/2023] Open
Abstract
Albumin is an attractive candidate carrier for the development of novel therapeutic drugs. Gemcitabine has been FDA approved for the treatment of solid tumors; however, new drugs that optimize gemcitabine delivery are not available for clinical use. The aim of this study was to test the efficacy of a novel albumin-encapsulated gemcitabine prodrug, JNTX-101, and investigate whether Cav-1 expression predicts the therapeutic efficacy of JNTX-101. We first determined the treatment efficacy of JNTX-101 in a panel of pancreatic/lung cancer cell lines and found that increases in Cav-1 expression resulted in higher uptake of albumin, while Cav-1 depletion attenuated the sensitivity of cells to JNTX-101. In addition, decreased Cav-1 expression markedly reduced JNTX-101-induced apoptotic cell death in a panel of cells, particularly in low-serum conditions. Furthermore, we tested the therapeutic efficacy of JNTX-101 in xenograft models and the role of Cav-1 in JNTX-101 sensitivity using a Tet-on-inducible tumor model in vivo. Our data suggest that JNTX-101 effectively inhibits cell viability and tumor growth, and that Cav-1 expression dictates optimal sensitivity to JNTX-101. These data indicate that Cav-1 correlates with JNTX-101 sensitivity, especially under nutrient-deprived conditions, and supports a role for Cav-1 as a predictive biomarker for albumin-encapsulated therapeutics such as JNTX-101.
Collapse
Affiliation(s)
- Tiantian Cui
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | | | | | - Sindhu Nair
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | | | | | | | - Sam Ellis
- January Therapeutics, San Diego, CA 92121, USA
| | | | - Neil Raheja
- January Therapeutics, San Diego, CA 92121, USA
| | - Terence M. Williams
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
46
|
Hu H, Ng TS, Kang M, Scott E, Li R, Quintana JM, Matvey D, Vantaku VR, Weissleder R, Parangi S, Miller MA. Thyroid Cancers Exhibit Oncogene-Enhanced Macropinocytosis that Is Restrained by IGF1R and Promote Albumin-Drug Conjugate Response. Clin Cancer Res 2023; 29:3457-3470. [PMID: 37289199 PMCID: PMC10527034 DOI: 10.1158/1078-0432.ccr-22-2976] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/22/2022] [Accepted: 06/06/2023] [Indexed: 06/09/2023]
Abstract
PURPOSE Oncogene-driven macropinocytosis fuels nutrient scavenging in some cancer types, yet whether this occurs in thyroid cancers with prominent MAPK-ERK and PI3K pathway mutations remains unclear. We hypothesized that understanding links between thyroid cancer signaling and macropinocytosis might uncover new therapeutic strategies. EXPERIMENTAL DESIGN Macropinocytosis was assessed across cells derived from papillary thyroid cancer (PTC), follicular thyroid cancer (FTC), non-malignant follicular thyroid, and aggressive anaplastic thyroid cancer (ATC), by imaging fluorescent dextran and serum albumin. The impacts of ectopic BRAFV600E and mutant RAS, genetic PTEN silencing, and inhibitors targeting RET, BRAF, and MEK kinases were quantified. BrafV600E p53-/- ATC tumors in immunocompetent mice were used to measure efficacy of an albumin-drug conjugate comprising microtubule-destabilizing monomethyl auristatin E (MMAE) linked to serum albumin via a cathepsin-cleavable peptide (Alb-vc-MMAE). RESULTS FTC and ATC cells showed greater macropinocytosis than non-malignant and PTC cells. ATC tumors accumulated albumin at 8.8% injected dose per gram tissue. Alb-vc-MMAE, but not MMAE alone, reduced tumor size by >90% (P < 0.01). ATC macropinocytosis depended on MAPK/ERK activity and nutrient signaling, and increased by up to 230% with metformin, phenformin, or inhibition of IGF1Ri in monoculture but not in vivo. Macrophages also accumulated albumin and express the cognate IGF1R ligand, IGF1, which reduced ATC responsiveness to IGF1Ri. CONCLUSIONS These findings identify regulated oncogene-driven macropinocytosis in thyroid cancers and demonstrate the potential of designing albumin-bound drugs to efficiently treat them.
Collapse
Affiliation(s)
- Huiyu Hu
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
- Department of General Surgery, Xiangya Hospital, Central South University, China
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Mikyung Kang
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Ella Scott
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Jeremy M. Quintana
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Dylan Matvey
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
| | - Venkata R. Vantaku
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
- Department of Systems Biology, Harvard Medical School, United States
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| |
Collapse
|
47
|
Zhao DK, Liang J, Huang XY, Shen S, Wang J. Organoids technology for advancing the clinical translation of cancer nanomedicine. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1892. [PMID: 37088100 DOI: 10.1002/wnan.1892] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 04/25/2023]
Abstract
The past decades have witnessed the rapid development and widespread application of nanomedicines in cancer treatment; however, the clinical translation of experimental findings has been low, as evidenced by the low percentage of commercialized nanomedicines. Incomplete understanding of nanomedicine-tumor interactions and inappropriate evaluation models are two important challenges limiting the clinical translation of cancer nanomedicines. Currently, nanomedicine-tumor interaction and therapeutic effects are mainly investigated using cell lines or mouse models, which do not recapitulate the complex tumor microenvironment in human patients. Thus, information obtained from cell lines and mouse models cannot provide adequate guidance for the rational redesign of nanomedicine. Compared with other preclinical models, tumor organoids constructed from patient-derived tumor tissues are superior in retaining the key histopathological, genetic, and phenotypic features of the parent tumor. We speculate that organoid technology would help elucidate nanomedicine-tumor interaction in the tumor microenvironment and guide the design of nanomedicine, making it a reliable tool to accurately predict drug responses in patients with cancer. This review highlighted the advantages of drug delivery systems in cancer treatment, challenges limiting the clinical translation of antitumor nanomedicines, and potential application of patient-derived organoids (PDO) in nanomedicine. We propose that combining organoids and nanotechnology would facilitate the development of safe and effective cancer nanomedicines and accelerate their clinical application. This review discussed the potential translational value of integrative research using organoids and cancer nanomedicine. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Dong-Kun Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Jie Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Xiao-Yi Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, China
| |
Collapse
|
48
|
Song C, Chen M, Tan J, Xu J, Zhang Y, Zhang G, Hu X, Liu S. Self-Amplified Cascade Degradation and Oxidative Stress Via Rational pH Regulation of Oxidation-Responsive Poly(ferrocene) Aggregates. J Am Chem Soc 2023; 145:17755-17766. [PMID: 37527404 DOI: 10.1021/jacs.3c04454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Precise activation of polymer nanoparticles at lesion sites is crucial to achieve favorable therapeutic efficacy. However, conventional endogenous stimuli-responsive polymer nanoparticles probably suffer from few triggers to stimulate the polymer degradation and subsequent functions. Here, we describe oxidation-responsive poly(ferrocene) amphiphiles containing phenylboronic acid ester and ferrocene as the repeating backbone units. Upon triggering by hydrogen peroxide inside the tumor cells, the phenylboronic acid ester bonds are broken and poly(ferrocene) units are degraded to afford free ferrocene and noticeable hydroxide ions. The released hydroxide ions can immediately improve the pH value within the poly(ferrocene) aggregates, and the degradation rate of the phenylboronic acid ester backbone is further promoted by the upregulated pH; thereupon, the accelerated degradation can release much more additional hydroxide ions to improve the pH, thus achieving a positive self-amplified cascade degradation of poly(ferrocene) aggregates accompanied by oxidative stress boosting and efficient cargo release. Specifically, the poly(ferrocene) aggregates can be degraded up to ∼90% within 12 h when triggered by H2O2, while ferrocene-free control nanoparticles are degraded by only 30% within 12 days. In addition, the maleimide moieties tethered in the hydrophilic corona can capture blood albumin to form an albumin-rich protein corona and significantly improve favorable tumor accumulation. The current oxidation-responsive poly(ferrocene) amphiphiles can efficiently inhibit tumors in vitro and in vivo. This work provides a proof-of-concept paradigm for self-amplified polymer degradation and concurrent oxidative stress, which is promising in actively regulated precision medicine.
Collapse
Affiliation(s)
- Chengzhou Song
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Minglong Chen
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Jiajia Tan
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Jie Xu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Yuben Zhang
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Guoying Zhang
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Xianglong Hu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, China
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Shiyong Liu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, China
| |
Collapse
|
49
|
Li C, Zhang D, Pan Y, Chen B. Human Serum Albumin Based Nanodrug Delivery Systems: Recent Advances and Future Perspective. Polymers (Basel) 2023; 15:3354. [PMID: 37631411 PMCID: PMC10459149 DOI: 10.3390/polym15163354] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
With the success of several clinical trials of products based on human serum albumin (HSA) and the rapid development of nanotechnology, HSA-based nanodrug delivery systems (HBNDSs) have received extensive attention in the field of nanomedicine. However, there is still a lack of comprehensive reviews exploring the broader scope of HBNDSs in biomedical applications beyond cancer therapy. To address this gap, this review takes a systematic approach. Firstly, it focuses on the crystal structure and the potential binding sites of HSA. Additionally, it provides a comprehensive summary of recent progresses in the field of HBNDSs for various biomedical applications over the past five years, categorized according to the type of therapeutic drugs loaded onto HSA. These categories include small-molecule drugs, inorganic materials and bioactive ingredients. Finally, the review summarizes the characteristics and current application status of HBNDSs in drug delivery, and also discusses the challenges that need to be addressed for the clinical transformation of HSA formulations and offers future perspectives in this field.
Collapse
Affiliation(s)
- Changyong Li
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China;
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China
| | - Dagui Zhang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China
| | - Yujing Pan
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China
| | - Biaoqi Chen
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China;
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China
| |
Collapse
|
50
|
Andrade F, German-Cortés J, Montero S, Carcavilla P, Baranda-Martínez-Abascal D, Moltó-Abad M, Seras-Franzoso J, Díaz-Riascos ZV, Rafael D, Abasolo I. The Nanotechnology-Based Approaches against Kirsten Rat Sarcoma-Mutated Cancers. Pharmaceutics 2023; 15:1686. [PMID: 37376135 DOI: 10.3390/pharmaceutics15061686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/18/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Kirsten rat sarcoma (KRAS) is a small GTPase which acts as a molecular switch to regulate several cell biological processes including cell survival, proliferation, and differentiation. Alterations in KRAS have been found in 25% of all human cancers, with pancreatic cancer (90%), colorectal cancer (45%), and lung cancer (35%) being the types of cancer with the highest mutation rates. KRAS oncogenic mutations are not only responsible for malignant cell transformation and tumor development but also related to poor prognosis, low survival rate, and resistance to chemotherapy. Although different strategies have been developed to specifically target this oncoprotein over the last few decades, almost all of them have failed, relying on the current therapeutic solutions to target proteins involved in the KRAS pathway using chemical or gene therapy. Nanomedicine can certainly bring a solution for the lack of specificity and effectiveness of anti-KRAS therapy. Therefore, nanoparticles of different natures are being developed to improve the therapeutic index of drugs, genetic material, and/or biomolecules and to allow their delivery specifically into the cells of interest. The present work aims to summarize the most recent advances related to the use of nanotechnology for the development of new therapeutic strategies against KRAS-mutated cancers.
Collapse
Affiliation(s)
- Fernanda Andrade
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Departament de Farmàcia i Tecnologia Farmacèutica i Fisicoquímica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Júlia German-Cortés
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
| | - Sara Montero
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
| | - Pilar Carcavilla
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
| | - Diego Baranda-Martínez-Abascal
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
| | - Marc Moltó-Abad
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Joaquín Seras-Franzoso
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain
| | - Zamira Vanessa Díaz-Riascos
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Diana Rafael
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Ibane Abasolo
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, 08035 Barcelona, Spain
- Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
- Clinical Biochemistry Service, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| |
Collapse
|