1
|
Van der Sanden N, Paun RA, Yitayew MY, Boyadjian O, Tabrizian M. An investigation of the effect of the protein corona on the cellular uptake of nanoliposomes under flow conditions using quartz crystal microgravimetry with dissipation. NANOSCALE ADVANCES 2024; 7:169-184. [PMID: 39569329 PMCID: PMC11575535 DOI: 10.1039/d4na00783b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/24/2024] [Indexed: 11/22/2024]
Abstract
When nanoparticle delivery systems are immersed in biological fluids, a complex assembly of proteins forms on their surface, creating a protein corona. The protein corona alters the physicochemical properties, toxicity, biodistribution, cellular uptake, and immune response of the nanoparticles, and consequently, their therapeutic efficacy. Currently, there is a lack of in vitro methods to assess the effects of the protein corona on nanoparticle uptake under dynamic flow and assess their binding kinetics in real-time. Here, we introduce quartz crystal microbalance with dissipation (QCM-D) as an in vitro technique, capable of incorporating dynamic flow, to study the effect of the protein corona on the binding of nanoliposome (NLP) formulations to cell surfaces as a first step in their cellular uptake. The interactions of four NLP formulations (low PEGylated, high PEGylated, negatively charged and positively charged NLPs) with A375 melanoma and THP1 cell lines were assessed by QCM-D, before and after the formation of a protein corona. Through real-time recording of the frequency and dissipation shifts (Δf and ΔD, respectively), the QCM-D results provided strong evidence of the role of the protein corona in the cellular interaction of these NLP formulations, with a variation in their adsorption kinetics depending on their initial composition. NLP's attachment to the cell surface was the lowest for PEGylated NLPs (<5%), while the positively charged NLPs showed the highest cellular attachment (≈100%), regardless of the presence of the protein corona or cell type. The effect of the protein corona was more pronounced for the negatively charged NLPs, where a significant reduction in the NLP attachment was observed. To complement the QCM-D data on the NLP attachment and to determine whether the NLP attachment leads to cellular uptake, confocal microscopy and flow cytometry were used to confirm NLP uptake by A375 and THP1 cells. Proteomic analysis revealed a differential composition of the protein corona on the various NLPs with possible implications for their sequestration and cellular uptake. Collectively, the findings suggest that QCM-D can be an important tool to study the binding of NLP formulations or other nanoparticles with cell membranes under dynamic flow, which very often differs from nanoparticle uptake under static conditions.
Collapse
Affiliation(s)
- Nicholas Van der Sanden
- Department of Biomedical Engineering, McGill University Duff Medical Building, 3775 University Street Montreal Quebec H3A 2B4 Canada
| | - Radu A Paun
- Department of Biomedical Engineering, McGill University Duff Medical Building, 3775 University Street Montreal Quebec H3A 2B4 Canada
| | - Michael Y Yitayew
- Department of Biomedical Engineering, McGill University Duff Medical Building, 3775 University Street Montreal Quebec H3A 2B4 Canada
| | - Oscar Boyadjian
- Department of Biomedical Engineering, McGill University Duff Medical Building, 3775 University Street Montreal Quebec H3A 2B4 Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University Duff Medical Building, 3775 University Street Montreal Quebec H3A 2B4 Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University Montreal Canada
| |
Collapse
|
2
|
Zhuang Z, Kong W, Wen Z, Tong N, Lin J, Zhang F, Fan Z, Yi L, Huang Y, Duan Y, Yan X, Zhu X. Combinatorial metabolic engineering of Streptomyces sp. CB03234-S for the enhanced production of anthraquinone-fused enediyne tiancimycins. Microb Cell Fact 2024; 23:128. [PMID: 38704580 PMCID: PMC11069151 DOI: 10.1186/s12934-024-02399-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Anthraquinone-fused enediynes (AFEs) are excellent payloads for antibody-drug conjugates (ADCs). The yields of AFEs in the original bacterial hosts are extremely low. Multiple traditional methods had been adopted to enhance the production of the AFEs. Despite these efforts, the production titers of these compounds are still low, presenting a practical challenge for their development. Tiancimycins (TNMs) are a class of AFEs produced by Streptomyces sp. CB03234. One of their salient features is that they exhibit rapid and complete cell killing ability against various cancer cell lines. RESULTS In this study, a combinatorial metabolic engineering strategy guided by the CB03234-S genome and transcriptome was employed to improve the titers of TNMs. First, re-sequencing of CB03234-S (Ribosome engineered mutant strains) genome revealed the deletion of a 583-kb DNA fragment, accounting for about 7.5% of its genome. Second, by individual or combined inactivation of seven potential precursor competitive biosynthetic gene clusters (BGCs) in CB03234-S, a double-BGC inactivation mutant, S1009, was identified with an improved TNMs titer of 28.2 ± 0.8 mg/L. Third, overexpression of five essential biosynthetic genes, including two post-modification genes, and three self-resistance auxiliary genes, was also conducted, through which we discovered that mutants carrying the core genes, tnmE or tnmE10, exhibited enhanced TNMs production. The average TNMs yield reached 43.5 ± 2.4 mg/L in a 30-L fermenter, representing an approximately 360% increase over CB03234-S and the highest titer among all AFEs to date. Moreover, the resulting mutant produced TNM-W, a unique TNM derivative with a double bond instead of a common ethylene oxide moiety. Preliminary studies suggested that TNM-W was probably converted from TNM-A by both TnmE and TnmE10. CONCLUSIONS Based on the genome and transcriptome analyses, we adopted a combined metabolic engineering strategy for precursor enrichment and biosynthetic pathway reorganization to construct a high-yield strain of TNMs based on CB03234-S. Our study establishes a solid basis for the clinical development of AFE-based ADCs.
Collapse
Affiliation(s)
- Zhoukang Zhuang
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China
| | - Wenping Kong
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China
| | - Zhongqing Wen
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China
| | - Nian Tong
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China
| | - Jing Lin
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China
| | - Fan Zhang
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China
| | - Zhiying Fan
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China
| | - Liwei Yi
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China
- The Affiliated Nanhua Hospital, Department of Pharmacy, Institute of Clinical Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421002, China
| | - Yong Huang
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug Discovery, Changsha, 410011, China
| | - Yanwen Duan
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China.
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug Discovery, Changsha, 410011, China.
- National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha, 410013, China.
| | - Xiaohui Yan
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China.
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Xiangcheng Zhu
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, 410013, China.
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug Discovery, Changsha, 410011, China.
- National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha, 410013, China.
| |
Collapse
|
3
|
Feng X, Wen Z, Zhu X, Yan X, Duan Y, Huang Y. Anti-HER2 Immunoliposomes: Antitumor Efficacy Attributable to Targeted Delivery of Anthraquinone-Fused Enediyne. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307865. [PMID: 38355309 PMCID: PMC11077693 DOI: 10.1002/advs.202307865] [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: 10/18/2023] [Revised: 12/24/2023] [Indexed: 02/16/2024]
Abstract
Although natural products are essential sources of small-molecule antitumor drugs, some can exert substantial toxicities, limiting their clinical utility. Anthraquinone-fused enediyne natural products are remarkably potent antitumor drug candidates, and uncialamycin and tiancimycin (TNM) A are under development as antibody-drug conjugates. Herein, a novel drug delivery system is introduced for TNM A using anti-human epidermal growth factor receptor 2 (HER2) immunoliposomes (ILs). Trastuzumab-coated TNM A-loaded ILs (HER2-TNM A-ILs) is engineered with an average particle size of 182.8 ± 2.1 nm and a zeta potential of 1.75 ± 0.12 mV. Compared with liposomes lacking trastuzumab, HER2-TNM A-ILs exhibited selective toxicity against HER2-positive KPL-4 and SKBR3 cells. Coumarin-6, a fluorescent TNM A surrogate, is encapsulated within anti-HER2 ILs; the resultant ILs have enhanced cellular uptake in KPL-4 and SKBR3 cells when compared with control liposomes. Furthermore, ILs loaded with more Cy5.5 accumulated in KPL-4 mouse tumors. A single HER2-TNM A-IL dose (0.02 mg kg-1) suppressed the growth of HER2-positive KPL-4 mouse tumors without apparent toxicity. This study not only provides a straightforward method for the effective delivery of TNM A against HER2-positive breast tumors but also underscores the potential of IL-based drug delivery systems when employing highly potent cytotoxins as payloads.
Collapse
Affiliation(s)
- Xueqiong Feng
- Xiangya International Academy of Translational MedicineCentral South UniversityChangshaHunan410013China
| | - Zhongqing Wen
- Xiangya International Academy of Translational MedicineCentral South UniversityChangshaHunan410013China
| | - Xiangcheng Zhu
- Xiangya International Academy of Translational MedicineCentral South UniversityChangshaHunan410013China
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug DiscoverChangshaHunan410011China
| | - Xiaohui Yan
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Yanwen Duan
- Xiangya International Academy of Translational MedicineCentral South UniversityChangshaHunan410013China
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug DiscoverChangshaHunan410011China
- National Engineering Research Center of Combinatorial Biosynthesis for Drug DiscoveryChangshaHunan410011China
| | - Yong Huang
- Xiangya International Academy of Translational MedicineCentral South UniversityChangshaHunan410013China
- National Engineering Research Center of Combinatorial Biosynthesis for Drug DiscoveryChangshaHunan410011China
- Institute of Health and MedicineHefei Comprehensive National Science CenterHefeiAnhui230093China
| |
Collapse
|
4
|
Yan H, Xu P, Cong H, Yu B, Shen Y. Research progress in construction of organic carrier drug delivery platform using tumor microenvironment. MATERIALS TODAY CHEMISTRY 2024; 37:101997. [DOI: 10.1016/j.mtchem.2024.101997] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Wen Z, Zhuang Z, Liu H, Wang Z, Feng X, Zhu X, Yan X, Duan Y, Huang Y. DNA Interaction and Cleavage Modes of Anthraquinone-Fused Enediynes: A Study on Tiancimycins, Yangpumicins, and Their Semisynthetic Analogues. J Med Chem 2024; 67:4624-4640. [PMID: 38483132 DOI: 10.1021/acs.jmedchem.3c02049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Dynemicin A has been the sole prototypical anthraquinone-fused enediyne (AFE) explored since its discovery in 1989. This study investigates the distinct DNA binding and cleavage mechanisms of emerging AFEs, represented by tiancimycins and yangpumicins, along with semisynthetic analogues. Our findings reveal their potent cytotoxicity against various tumor cell lines, while 18-methoxy tiancimycin A treatment could significantly suppress breast tumor growth with minimal toxicity. One of the most potent AFEs, i.e., tiancimycin A, preferentially targets DNA sequences 5'-ATT, 5'-CTT, 5'-GAA, 5'-GAT, and 5'-TTA. Molecular dynamics simulations suggest that emerging AFEs intercalate deeper into AT-rich DNA base pairs compared to dynemicin A. Importantly, tiancimycin A may equilibrate between insertional and intercalative modes without deintercalation, enabling selective cleavage of T and A bases. This study underscores how subtle structural variations among AFEs significantly influence their DNA recognition and cleavage, facilitating future design of novel AFEs as potent and selective payloads for antibody-drug conjugates.
Collapse
Affiliation(s)
- Zhongqing Wen
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
| | - Zhoukang Zhuang
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
| | - Huiming Liu
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
| | - Zilong Wang
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
| | - Xueqiong Feng
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
| | - Xiangcheng Zhu
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug Discovery, Changsha, Hunan 410011, China
| | - Xiaohui Yan
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanwen Duan
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug Discovery, Changsha, Hunan 410011, China
- National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha, Hunan 410011, China
| | - Yong Huang
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
- National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha, Hunan 410011, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| |
Collapse
|
6
|
Sun EG, Vijayan V, Park MR, Yoo KH, Cho SH, Bae WK, Shim HJ, Hwang JE, Park IK, Chung IJ. Suppression of triple-negative breast cancer aggressiveness by LGALS3BP via inhibition of the TNF-α-TAK1-MMP9 axis. Cell Death Discov 2023; 9:122. [PMID: 37041137 PMCID: PMC10090165 DOI: 10.1038/s41420-023-01419-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/13/2023] Open
Abstract
Transforming growth factor-β-activated kinase 1 (TAK1), which is highly expressed and aberrantly activated in triple-negative breast cancer (TNBC), plays a pivotal role in metastasis and progression. This makes it a potential therapeutic target for TNBC. Previously, we reported lectin galactoside-binding soluble 3 binding protein (LGALS3BP) as a negative regulator of TAK1 signaling in the inflammatory response and inflammation-associated cancer progression. However, the role of LGALS3BP and its molecular interaction with TAK1 in TNBC remain unclear. This study aimed to investigate the function and underlying mechanism of action of LGALS3BP in TNBC progression and determine the therapeutic potential of nanoparticle-mediated delivery of LGALS3BP in TNBC. We found that LGALS3BP overexpression suppressed the overall aggressive phenotype of TNBC cells in vitro and in vivo. LGALS3BP inhibited TNF-α-mediated gene expression of matrix metalloproteinase 9 (MMP9), which encodes a protein crucial for lung metastasis in TNBC patients. Mechanistically, LGALS3BP suppressed TNF-α-mediated activation of TAK1, a key kinase linking TNF-α stimulation and MMP9 expression in TNBC. Nanoparticle-mediated delivery enabled tumor-specific targeting and inhibited TAK1 phosphorylation and MMP9 expression in tumor tissues, suppressing primary tumor growth and lung metastasis in vivo. Our findings reveal a novel role of LGALS3BP in TNBC progression and demonstrate the therapeutic potential of nanoparticle-mediated delivery of LGALS3BP in TNBC.
Collapse
Affiliation(s)
- Eun-Gene Sun
- Department of Hematology and Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
- Immunotherapy Innovation Center, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Veena Vijayan
- Department of Biomedical Sciences and Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Mi-Ra Park
- Department of Hematology and Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Kyung Hyun Yoo
- Department of Biological Science, Sookmyung Women's University, Seoul, Republic of Korea
| | - Sang-Hee Cho
- Department of Hematology and Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
- Immunotherapy Innovation Center, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Woo-Kyun Bae
- Department of Hematology and Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC Center, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Hyun-Jeong Shim
- Department of Hematology and Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Jun-Eul Hwang
- Department of Hematology and Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences and Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju, Republic of Korea.
| | - Ik-Joo Chung
- Department of Hematology and Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea.
- Immunotherapy Innovation Center, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea.
| |
Collapse
|
7
|
Prajapat VM, Mahajan S, Paul PG, Aalhate M, Mehandole A, Madan J, Dua K, Chellappan DK, Singh SK, Singh PK. Nanomedicine: A pragmatic approach for tackling melanoma skin cancer. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
8
|
Advances in the Application of Nanomaterials to the Treatment of Melanoma. Pharmaceutics 2022; 14:pharmaceutics14102090. [PMID: 36297527 PMCID: PMC9610396 DOI: 10.3390/pharmaceutics14102090] [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: 08/03/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022] Open
Abstract
Melanoma can be divided into cutaneous melanoma, uveal melanoma, mucosal melanoma, etc. It is a very aggressive tumor that is prone to metastasis. Patients with metastatic melanoma have a poor prognosis and shorter survival. Although current melanoma treatments have been dramatically improved, there are still many problems such as systemic toxicity and the off-target effects of drugs. The use of nanoparticles may overcome some inadequacies of current melanoma treatments. In this review, we summarize the limitations of current therapies for cutaneous melanoma, uveal melanoma, and mucosal melanoma, as well as the adjunct role of nanoparticles in different treatment modalities. We suggest that nanomaterials may have an effective intervention in melanoma treatment in the future.
Collapse
|
9
|
Xu S, Zhang X, Zhu X, Su H, Yan X. A combined arsenic trioxide/tetrandrine nanoparticle formulation with improved inhibitory effect against promyelocytic leukemia. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|