1
|
Cai X, Xu W, Ren C, Zhang L, Zhang C, Liu J, Yang C. Recent progress in quantitative analysis of self-assembled peptides. EXPLORATION (BEIJING, CHINA) 2024; 4:20230064. [PMID: 39175887 PMCID: PMC11335468 DOI: 10.1002/exp.20230064] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/05/2023] [Indexed: 08/24/2024]
Abstract
Self-assembled peptides have been among the important biomaterials due to its excellent biocompatibility and diverse functions. Over the past decades, substantial progress and breakthroughs have been made in designing self-assembled peptides with multifaceted biomedical applications. The techniques for quantitative analysis, including imaging-based quantitative techniques, chromatographic technique and computational approach (molecular dynamics simulation), are becoming powerful tools for exploring the structure, properties, biomedical applications, and even supramolecular assembly processes of self-assembled peptides. However, a comprehensive review concerning these quantitative techniques remains scarce. In this review, recent progress in techniques for quantitative investigation of biostability, cellular uptake, biodistribution, self-assembly behaviors of self-assembled peptide etc., are summarized. Specific applications and roles of these techniques are highlighted in detail. Finally, challenges and outlook in this field are concluded. It is believed that this review will provide technical guidance for researchers in the field of peptide-based materials and pharmaceuticals, and facilitate related research for newcomers in this field.
Collapse
Affiliation(s)
- Xiaoyao Cai
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Wei Xu
- Department of PathologyCharacteristic Medical Center of Chinese People's Armed Police ForcesTianjinP. R. China
| | - Chunhua Ren
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Liping Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Congrou Zhang
- Metabolomics and Analytics Center, Leiden Academic Centre of Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Cuihong Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| |
Collapse
|
2
|
Chen Q, Yuan L, Chou WC, Cheng YH, He C, Monteiro-Riviere NA, Riviere JE, Lin Z. Meta-Analysis of Nanoparticle Distribution in Tumors and Major Organs in Tumor-Bearing Mice. ACS NANO 2023; 17:19810-19831. [PMID: 37812732 PMCID: PMC10604101 DOI: 10.1021/acsnano.3c04037] [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: 05/05/2023] [Accepted: 08/24/2023] [Indexed: 10/11/2023]
Abstract
Low tumor delivery efficiency is a critical barrier in cancer nanomedicine. This study reports an updated version of "Nano-Tumor Database", which increases the number of time-dependent concentration data sets for different nanoparticles (NPs) in tumors from the previous version of 376 data sets with 1732 data points from 200 studies to the current version of 534 data sets with 2345 data points from 297 studies published from 2005 to 2021. Additionally, the current database includes 1972 data sets for five major organs (i.e., liver, spleen, lung, heart, and kidney) with a total of 8461 concentration data points. Tumor delivery and organ distribution are calculated using three pharmacokinetic parameters, including delivery efficiency, maximum concentration, and distribution coefficient. The median tumor delivery efficiency is 0.67% injected dose (ID), which is low but is consistent with previous studies. Employing the best regression model for tumor delivery efficiency, we generate hypothetical scenarios with different combinations of NP factors that may lead to a higher delivery efficiency of >3%ID, which requires further experimentation to confirm. In healthy organs, the highest NP accumulation is in the liver (10.69%ID/g), followed by the spleen 6.93%ID/g and the kidney 3.22%ID/g. Our perspective on how to facilitate NP design and clinical translation is presented. This study reports a substantially expanded "Nano-Tumor Database" and several statistical models that may help nanomedicine design in the future.
Collapse
Affiliation(s)
- Qiran Chen
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| | - Long Yuan
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| | - Wei-Chun Chou
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| | - Yi-Hsien Cheng
- Department
of Anatomy and Physiology, Kansas State
University, Manhattan, Kansas 66506, United States
- Institute
of Computational Comparative Medicine, Kansas
State University, Manhattan, Kansas 66506, United States
| | - Chunla He
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Department
of Biostatistics College of Public Health and Health Professions, University of Florida, Gainesville, Florida 32608, United States
| | - Nancy A. Monteiro-Riviere
- Nanotechnology
Innovation Center of Kansas State, Kansas
State University, Manhattan, Kansas 66506, United States
- Center
for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Jim E. Riviere
- Center
for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, North Carolina 27606, United States
- 1
Data Consortium, Kansas State University, Olathe, Kansas 66061, United States
| | - Zhoumeng Lin
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
3
|
Horie S, Suzuki Y, Yamamoto T, Obika S, Mohri K, Kiyota C, Ren Q, Warashina S, Wada Y, Watanabe Y, Mukai H, Sato Y. Novel strategy of liver cancer treatment with modified antisense oligonucleotides targeting human vasohibin-2. Cancer Sci 2023; 114:3740-3749. [PMID: 37430466 PMCID: PMC10475766 DOI: 10.1111/cas.15897] [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: 04/05/2023] [Revised: 05/16/2023] [Accepted: 06/14/2023] [Indexed: 07/12/2023] Open
Abstract
Vasohihibin-2 (VASH2) is a homolog of vasohibin-1 (VASH1) and is overexpressed in various cancers. Vasohihibin-2 acts on both cancer cells and cancer microenvironmental cells. Previous analyses have shown that VASH2 promotes cancer progression and abrogation of VASH2 results in significant anticancer effects. We therefore propose VASH2 to be a practical molecular target for cancer treatment. Modifications of antisense oligonucleotide (ASO) such as bridged nucleic acids (BNA)-based modification increases the specificity and stability of ASO, and are now applied to the development of a number of oligonucleotide-based drugs. Here we designed human VASH2-ASOs, selected an optimal one, and developed 2',4'-BNA-based VASH2-ASO. When systemically administered, naked 2',4'-BNA-based VASH2-ASO accumulated in the liver and showed its gene-silencing activity. We then examined the effect of 2',4'-BNA-based VASH2-ASO in liver cancers. Intraperitoneal injection of naked 2',4'-BNA-based VASH2-ASO exerted a potent antitumor effect on orthotopically inoculated human hepatocellular carcinoma cells. The same manipulation also showed potent antitumor activity on the splenic inoculation of human colon cancer cells for liver metastasis. These results provide a novel strategy for the treatment of primary as well as metastatic liver cancers by using modified ASOs targeting VASH2.
Collapse
Affiliation(s)
- Sachiko Horie
- Department of Vascular BiologyInstitute of Development, Aging and Cancer, Tohoku UniversitySendaiJapan
| | - Yasuhiro Suzuki
- Department of Vascular BiologyInstitute of Development, Aging and Cancer, Tohoku UniversitySendaiJapan
- New Industry Creation Hatchery CenterTohoku UniversitySendaiJapan
| | - Tsuyoshi Yamamoto
- Graduate School of Pharmaceutical SciencesOsaka UniversityOsakaJapan
- Present address:
Department of Chemistry of Biofunctional Molecules, School of Pharmaceutical SciencesNagasaki UniversityNagasakiJapan
| | - Satoshi Obika
- Graduate School of Pharmaceutical SciencesOsaka UniversityOsakaJapan
| | - Kohta Mohri
- Laboratory for Molecular Delivery and Imaging TechnologyRIKEN Center for Biosystems Dynamics ResearchKobeJapan
| | - Chizuru Kiyota
- Laboratory for Molecular Delivery and Imaging TechnologyRIKEN Center for Biosystems Dynamics ResearchKobeJapan
| | - Qin Ren
- Laboratory for Molecular Delivery and Imaging TechnologyRIKEN Center for Biosystems Dynamics ResearchKobeJapan
| | - Shota Warashina
- Laboratory for Molecular Delivery and Imaging TechnologyRIKEN Center for Biosystems Dynamics ResearchKobeJapan
| | - Yasuhiro Wada
- Laboratory for Pathophysiological and Health ScienceRIKEN Center for Biosystems Dynamics ResearchKobeJapan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health ScienceRIKEN Center for Biosystems Dynamics ResearchKobeJapan
| | - Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging TechnologyRIKEN Center for Biosystems Dynamics ResearchKobeJapan
- Department of Pharmaceutical Informatics, Graduate School of Biomedical SciencesNagasaki UniversityNagasakiJapan
| | - Yasufumi Sato
- Department of Vascular BiologyInstitute of Development, Aging and Cancer, Tohoku UniversitySendaiJapan
- New Industry Creation Hatchery CenterTohoku UniversitySendaiJapan
| |
Collapse
|
4
|
Abuhelal S, Centelles MN, Wright M, Mason AJ, Thanou M. Development of Cationic Lipid LAH4-L1 siRNA Complexes for Focused Ultrasound Enhanced Tumor Uptake. Mol Pharm 2023; 20:2341-2351. [PMID: 36989421 PMCID: PMC10155207 DOI: 10.1021/acs.molpharmaceut.2c00909] [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: 03/31/2023]
Abstract
RNAi has considerable potential as a cancer therapeutic approach, but effective and efficient delivery of short interfering RNA (siRNA) to tumors remains a major hurdle. It remains a challenge to prepare a functional siRNA complex, target enough dose to the tumor, and stimulate its internalization into tumor cells and its release to the cytoplasm. Here, we show how these key barriers to siRNA delivery can be overcome with a complex─comprising siRNA, cationic lipids, and pH-responsive peptides─that is suited to tumor uptake enhancement via focused ultrasound (FUS). The complex provides effective nucleic acid encapsulation, nuclease protection, and endosomal escape such that gene silencing in cells is substantially more effective than that obtained with either equivalent lipoplexes or commercial reagents. In mice bearing MDA-MB-231 breast cancer xenografts, both lipid and ternary, lipid:peptide:siRNA complexes, prepared with near-infrared fluorescently labeled siRNA, accumulate in tumors following FUS treatments. Therefore, combining a well-designed lipid:peptide:siRNA complex with FUS tumor treatments is a promising route to achieve robust in vivo gene delivery.
Collapse
Affiliation(s)
- Shahd Abuhelal
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Miguel N Centelles
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Michael Wright
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - A James Mason
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Maya Thanou
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| |
Collapse
|
5
|
Warashina S, Sato H, Zouda M, Takahashi M, Wada Y, Passioura T, Suga H, Watanabe Y, Matsumoto K, Mukai H. Two-Chain Mature Hepatocyte Growth Factor-Specific Positron Emission Tomography Imaging in Tumors Using 64Cu-Labeled HiP-8, a Nonstandard Macrocyclic Peptide Probe. Mol Pharm 2023; 20:2029-2038. [PMID: 36862642 DOI: 10.1021/acs.molpharmaceut.2c01020] [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: 03/03/2023]
Abstract
Two-chain hepatocyte growth factor (tcHGF), the mature form of HGF, is associated with malignancy and anticancer drug resistance; therefore, its quantification is an important indicator for cancer diagnosis. In tumors, activated tcHGF hardly discharges into the systemic circulation, indicating that tcHGF is an excellent target for molecular imaging using positron emission tomography (PET). We recently discovered HGF-inhibitory peptide-8 (HiP-8) that binds specifically to human tcHGF with nanomolar affinity. The purpose of this study was to investigate the usefulness of HiP-8-based PET probes in human HGF knock-in humanized mice. 64Cu-labeled HiP-8 molecules were synthesized using a cross-bridged cyclam chelator, CB-TE1K1P. Radio-high-performance liquid chromatography-based metabolic stability analyses showed that more than 90% of the probes existed in intact form in blood at least for 15 min. In PET studies, significantly selective visualization of hHGF-overexpressing tumors versus hHGF-negative tumors was observed in double-tumor-bearing mice. The accumulation of labeled HiP-8 into the hHGF-overexpressing tumors was significantly reduced by competitive inhibition. In addition, the radioactivity and distribution of phosphorylated MET/HGF receptor were colocalized in tissues. These results demonstrate that the 64Cu-labeled HiP-8 probes are suitable for tcHGF imaging in vivo, and secretory proteins like tcHGF can be a target for PET imaging.
Collapse
Affiliation(s)
- Shota Warashina
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hiroki Sato
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Maki Zouda
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Maiko Takahashi
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuhiro Wada
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Toby Passioura
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo 113-0033, Japan
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo 113-0033, Japan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan.,WPI-Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma, Kanazawa, Ishikawa 920-1192, Japan
| | - Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,Department of Pharmaceutical Informatics, Graduate School of Biomedical Science, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
6
|
Multimodal imaging distribution assessment of a liposomal antibiotic in an infectious disease model. J Control Release 2022; 352:199-210. [PMID: 36084816 DOI: 10.1016/j.jconrel.2022.08.061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/09/2022] [Accepted: 08/31/2022] [Indexed: 11/08/2022]
Abstract
Liposomes are promising targeted drug delivery systems with the potential to improve the efficacy and safety profile of certain classes of drugs. Though attractive, there are unique analytical challenges associated with the development of liposomal drugs including human dose prediction given these are multi-component drug delivery systems. In this study, we developed a multimodal imaging approach to provide a comprehensive distribution assessment for an antibacterial drug, GSK2485680, delivered as a liposomal formulation (Lipo680) in a mouse thigh model of bacterial infection to support human dose prediction. Positron emission tomography (PET) imaging was used to track the in vivo biodistribution of Lipo680 over 48 h post-injection providing a clear assessment of the uptake in various tissues and, importantly, the selective accumulation at the site of infection. In addition, a pharmacokinetic model was created to evaluate the kinetics of Lipo680 in different tissues. Matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) was then used to quantify the distribution of GSK2485680 and to qualitatively assess the distribution of a liposomal lipid throughout sections of infected and non-infected hindlimb tissues at high spatial resolution. Through the combination of both PET and MALDI IMS, we observed excellent correlation between the Lipo680-radionuclide signal detected by PET with the GSK2485680 and lipid component signals detected by MALDI IMS. This multimodal translational method can reduce drug attrition by generating comprehensive biodistribution profiles of drug delivery systems to provide mechanistic insight and elucidate safety concerns. Liposomal formulations have potential to deliver therapeutics across a broad array of different indications, and this work serves as a template to aid in delivering future liposomal drugs to the clinic.
Collapse
|
7
|
Tao W, Wang S, Xu A, Xue Y, Wang H, Xu H. 18F-FDG Micro PET/CT imaging to evaluate the effect of BRCA1 knockdown on MDA-MB231 breast cancer cell radiosensitivity. Transl Oncol 2022; 25:101517. [PMID: 35985202 PMCID: PMC9411683 DOI: 10.1016/j.tranon.2022.101517] [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: 06/08/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 12/04/2022] Open
Abstract
BRCA1 gene knockdown improves the radiosensitivity of breast cancer cells. BRCA1 gene knockdown combination with radiotherapy downregulates multiple biomarkers of poor prognosis. 18F-FDG Micro PET/CT imaging was able to evaluate the radiosensitizing effect of the BRCA1 gene in vitro experiment.
Objective Radioresistance of tumor cells is a major factor associated with failure of radiotherapy (RT). This study aimed to investigate the effect of BRCA1 knockdown on MDA-MB231 breast cancer cell radiosensitivity. Materials and methods Short hairpin RNA (shRNA) was used to knockdown BRCA1 gene in MDA-MB231 cells. Cell viability and proliferative capacity were assessed by CCK-8 and colony formation assays, respectively. We established xenograft models in nude mice to evaluate tumor volume and tumor weight. The mice were imaged by 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) before and after RT to evaluate changes in maximum standardized uptake value (SUVmax) and tumor SUVmax/muscle SUVmax (TMR). Changes in HIF-1α, Glut-1 and Ki-67 were analyzed and the correlation between 18F-FDG uptake and tumor biology was analyzed. Results Compared with the control cells, RT significantly reduced cell viability and colony formation capacity in cells with the BRCA1 gene knockdown. In vivo assays showed that there was obvious delay in the tumor growth in the shBRCA1+RT group compared with the control group. 18F-FDG Micro PET/CT indicated a reduction in glucose metabolism in the shBRCA1+RT group, with statistically significant differences in both the SUVmax and TMR. The data showed the expression of HIF-1α, Glut-1 and Ki-67 was downregulated in the shBRCA1+RT group, and both SUVmax and TMR had significant correlation with tumor biology. Conclusion These results demonstrated that BRCA1 knockdown improves the sensitivity of MDA-MB231 breast cancer cells to RT. In addition, 18F-FDG PET/CT imaging allows non-invasive analysis of tumor biology and assessment of radiosensitivity.
Collapse
Affiliation(s)
- Weitao Tao
- School of Basic Medical Sciences, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China
| | - Siqi Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Alei Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Yangyang Xue
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Hui Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Huiqin Xu
- School of Basic Medical Sciences, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China.
| |
Collapse
|
8
|
Wang Z, Li J, Lin G, He Z, Wang Y. Metal complex-based liposomes: Applications and prospects in cancer diagnostics and therapeutics. J Control Release 2022; 348:1066-1088. [PMID: 35718211 DOI: 10.1016/j.jconrel.2022.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 06/09/2022] [Indexed: 12/17/2022]
Abstract
Metal complexes are of increasing interest as pharmaceutical agents in cancer diagnostics and therapeutics, while some of them suffer from issues such as limited water solubility and severe systemic toxicity. These drawbacks severely hampered their efficacy and clinical applications. Liposomes hold promise as delivery vehicles for constructing metal complex-based liposomes to maximize the therapeutic efficacy and minimize the side effects of metal complexes. This review provides an overview on the latest advances of metal complex-based liposomal delivery systems. First, the development of metal complex-mediated liposomal encapsulation is briefly introduced. Next, applications of metal complex-based liposomes in a variety of fields are overviewed, where drug delivery, cancer imaging (single photon emission computed tomography (SPECT), positron emission tomography (PET), and magnetic resonance imaging (MRI)), and cancer therapy (chemotherapy, phototherapy, and radiotherapy) were involved. Moreover, the potential toxicity, action of toxic mechanisms, immunological effects of metal complexes as well as the advantages of metal complex-liposomes in this content are also discussed. In the end, the future expectations and challenges of metal complex-based liposomes in clinical cancer therapy are tentatively proposed.
Collapse
Affiliation(s)
- Zhaomeng Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Guimei Lin
- School of Pharmacy, Shandong University, Jinan 250000, PR China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
9
|
64Cu-labeling of small extracellular vesicle surfaces via a cross-bridged macrocyclic chelator for pharmacokinetic study by positron emission tomography imaging. Int J Pharm 2022; 624:121968. [PMID: 35772573 DOI: 10.1016/j.ijpharm.2022.121968] [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: 04/20/2022] [Revised: 06/10/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022]
Abstract
We developed a method of labeling the surfaces of small extracellular vesicles (sEVs) with 64Cu using a cross-bridged, macrocyclic chelator (CB-TE1A1P) and applied to pharmacokinetics study with positron emission tomography (PET). After incubation in 20% plasma for 10 min, approximately a half of the 64Cu was desorbed from 64Cu-labeled sEVs purified by phosphate-buffered saline wash, suggesting partly weak interaction without coordinating to CB-TE1A1P. After subsequent purification with albumin, 64Cu desorption was greatly reduced, resulting in a radiochemical stability of 95.7%. Notably, labeling did not alter the physicochemical and biological properties of sEVs. After intravenous injection, 64Cu-labeled sEVs rapidly disappeared from the systemic blood circulation and accumulated mainly in the liver and spleen of macrophage-competent mice. In macrophage-depleted mice, 64Cu-labeled sEVs remained in the blood circulation for a longer period and gradually accumulated in the liver and spleen, suggesting mechanisms of hepatic and splenic accumulation other than macrophage-dependent phagocytosis. The comparison of tissue uptake clearance between macrophage-competent and macrophage-depleted mice suggests that macrophages contributed to 67% and 76% of sEV uptake in the liver and spleen, respectively. The application of this method in pharmacokinetics PET studies can be useful in preclinical and clinical research and the development of sEV treatment modalities.
Collapse
|
10
|
Mukai H, Ogawa K, Kato N, Kawakami S. Recent advances in lipid nanoparticles for delivery of nucleic acid, mRNA, and gene editing-based therapeutics. Drug Metab Pharmacokinet 2022; 44:100450. [PMID: 35381574 PMCID: PMC9363157 DOI: 10.1016/j.dmpk.2022.100450] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 12/26/2022]
Abstract
Lipid nanoparticles (LNPs) are becoming popular as a means of delivering therapeutics, including those based on nucleic acids and mRNA. The mRNA-based coronavirus disease 2019 vaccines are perfect examples to highlight the role played by drug delivery systems in advancing human health. The fundamentals of LNPs for the delivery of nucleic acid- and mRNA-based therapeutics, are well established. Thus, future research on LNPs will focus on addressing the following: expanding the scope of drug delivery to different constituents of the human body, expanding the number of diseases that can be targeted, and studying the change in the pharmacokinetics of LNPs under physiological and pathological conditions. This review article provides an overview of recent advances aimed at expanding the application of LNPs, focusing on the pharmacokinetics and advantages of LNPs. In addition, analytical techniques, library construction and screening, rational design, active targeting, and applicability to gene editing therapy have also been discussed.
Collapse
Affiliation(s)
- Hidefumi Mukai
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan; Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Koki Ogawa
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan
| | - Naoya Kato
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan.
| |
Collapse
|
11
|
Talap J, Zhao J, Shen M, Song Z, Zhou H, Kang Y, Sun L, Yu L, Zeng S, Cai S. Recent advances in therapeutic nucleic acids and their analytical methods. J Pharm Biomed Anal 2021; 206:114368. [PMID: 34571322 DOI: 10.1016/j.jpba.2021.114368] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/11/2022]
Abstract
Therapeutic nucleic acids are various chemically modified RNA or DNA with different functions, which mainly play roles at the gene level. Owing to its accurately targeting at pathogenic genes, nucleic acid based therapeutics have a wide range of application prospects. Recently, the improvement on chemical synthesis and delivery materials accelerated the development of therapeutic nucleic acids rapidly. Up to now, 17 nucleic acid based therapeutics approved by Food and Drug Administration (FDA) or European Medicines Agency (EMA). The development of therapeutics raised higher requirements for analytical methods, both in quality control and in clinical research. The first part of this review introduces different classes of therapeutic nucleic acids, including antisense oligonucleotide (ASO), RNA interference (RNAi) therapy, mRNA, aptamer and other classes which are under research. The second part reviews the therapeutic nucleic acids commercialized from 2019 to now. The third part discusses the analytical methods for nucleic acid based therapeutics, including liquid chromatography-based methods, capillary gel electrophoresis (CGE), hybridization enzyme-linked immunosorbent assay (ELISA) and other infrequently used methods. Finally, the advantages and shortcomings of these methods are summarized, and the future development of analysis methods are prospected.
Collapse
Affiliation(s)
- Jadera Talap
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jing Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Minzhe Shen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zihan Song
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hui Zhou
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yu Kang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lianli Sun
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lushan Yu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Cancer Center of Zhejiang University, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Hangzhou, China.
| | - Sheng Cai
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Cancer Center of Zhejiang University, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Hangzhou, China.
| |
Collapse
|
12
|
Wang P, Zhou Y, Richards AM. Effective tools for RNA-derived therapeutics: siRNA interference or miRNA mimicry. Theranostics 2021; 11:8771-8796. [PMID: 34522211 PMCID: PMC8419061 DOI: 10.7150/thno.62642] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/30/2021] [Indexed: 12/18/2022] Open
Abstract
The approval of the first small interfering RNA (siRNA) drug Patisiran by FDA in 2018 marks a new era of RNA interference (RNAi) therapeutics. MicroRNAs (miRNA), an important post-transcriptional gene regulator, are also the subject of both basic research and clinical trials. Both siRNA and miRNA mimics are ~21 nucleotides RNA duplexes inducing mRNA silencing. Given the well performance of siRNA, researchers ask whether miRNA mimics are unnecessary or developed siRNA technology can pave the way for the emergence of miRNA mimic drugs. Through comprehensive comparison of siRNA and miRNA, we focus on (1) the common features and lessons learnt from the success of siRNAs; (2) the unique characteristics of miRNA that potentially offer additional therapeutic advantages and opportunities; (3) key areas of ongoing research that will contribute to clinical application of miRNA mimics. In conclusion, miRNA mimics have unique properties and advantages which cannot be fully matched by siRNA in clinical applications. MiRNAs are endogenous molecules and the gene silencing effects of miRNA mimics can be regulated or buffered to ameliorate or eliminate off-target effects. An in-depth understanding of the differences between siRNA and miRNA mimics will facilitate the development of miRNA mimic drugs.
Collapse
Affiliation(s)
- Peipei Wang
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
- Department of Medicine, National University Health System, 119228 Singapore
| | - Yue Zhou
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
- Department of Medicine, National University Health System, 119228 Singapore
| | - Arthur M. Richards
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
- Department of Medicine, National University Health System, 119228 Singapore
- Christchurch Heart Institute, Department of Medicine, University of Otago Christchurch, New Zealand
| |
Collapse
|
13
|
Wang R, Zhang Z, Liu B, Xue J, Liu F, Tang T, Liu W, Feng F, Qu W. Strategies for the design of nanoparticles: starting with long-circulating nanoparticles, from lab to clinic. Biomater Sci 2021; 9:3621-3637. [PMID: 34008587 DOI: 10.1039/d0bm02221g] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Short half-life is one of the main causes of drug attrition in clinical development, which also leads to the failure of many leading compounds and hits to become drug candidates. Nowadays, nanomaterials have been applied to drug development to address this problem. In fact, the clinical application of nanoparticles (NPs) is severely limited due to their rapid elimination by the reticuloendothelial system (RES) in vivo. In this paper, we aim to summarize representative strategies on prolonging the circulation time for bridging the gap between excellent pharmaceutics and proper half-life and encourage clinical translation.
Collapse
Affiliation(s)
- Ruyi Wang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Zhongtao Zhang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Bowen Liu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Jingwei Xue
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, 271000, China and Taian City institute of Digestive Disease, Taian City Central Hospital, Taian, 271000, China
| | - Fulei Liu
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, 271000, China and Pharmaceutical Department, Taian City Central Hospital, Taian, 271000, China
| | - Tongzhong Tang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China and Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China. and Jiangsu Food and Pharmaceutical Science College, Huaian, 223003, China.
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
14
|
Mukai H, Watanabe Y. Review: PET imaging with macro- and middle-sized molecular probes. Nucl Med Biol 2021; 92:156-170. [PMID: 32660789 DOI: 10.1016/j.nucmedbio.2020.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022]
Abstract
Recent progress in radiolabeling of macro- and middle-sized molecular probes has been extending possibilities to use PET molecular imaging for dynamic application to drug development and therapeutic evaluation. Theranostics concept also accelerated the use of macro- and middle-sized molecular probes for sharpening the contrast of proper target recognition even the cellular types/subtypes and proper selection of the patients who should be treated by the same molecules recognition. Here, brief summary of the present status of immuno-PET, and then further development of advanced technologies related to immuno-PET, peptidic PET probes, and nucleic acids PET probes are described.
Collapse
Affiliation(s)
- Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
15
|
Nomura S, Takahashi M, Kato AH, Wada Y, Watanabe Y, Yamashita F, Mukai H. Biosorption-based 64Cu-labeling of bacteria for pharmacokinetic positron-emission tomography. Int J Pharm 2020; 590:119950. [PMID: 33027635 DOI: 10.1016/j.ijpharm.2020.119950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023]
Abstract
Biosorption-based bacterial 64Cu-labeling and its application in pharmacokinetic positron-emission tomography (PET) were investigated. Both gram-positive and gram-negative bacteria were efficiently labeled with [64Cu]Cu2+ ion in saline at room temperature within 5 min. The labeling ratio for Escherichia coli drastically decreased with trypsin pretreatment and the co-presence of excess Cu2+ ion, indicating the existence of specific Cu2+ binding sites on the E. coli cell surface. Washing with lysogeny broth medium was effective in purifying 64Cu-labeled E. coli for kinetic study; the labeling stability was approximately 90% in serum for 15 min. According to dynamic PET imaging in colon-26 tumor-bearing mice, 64Cu-labeled E. coli immediately disappeared from the blood circulation and primarily accumulated in the liver. In addition, transient pulmonary distribution was observed, being in a dose-dependently accelerated manner. Considering the simplicity and versatility of biosorption-based bacterial 64Cu-labeling without genetic modification, the early-phase pharmacokinetic PET with 64Cu-labeled bacteria is promising for assessing toxicological aspects of bacteria-mediated cancer therapy as well as a variety of bacterial pathogenicities in infectious diseases.
Collapse
Affiliation(s)
- Shoko Nomura
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachi cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Maiko Takahashi
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Akari Hashiba Kato
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuhiro Wada
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachi cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Department of Pharmaceutical Informatics, Graduate School of Biomedical Science, Nagasaki University, 1-7-1 Sakamotomachi, Nagasaki 852-8588, Japan.
| |
Collapse
|
16
|
Chu Y, Zhang Z, He Q, Chen F, Sheng Z, Zhang D, Jin H, Jiang F, Guo L. Half-life determination of inorganic-organic hybrid nanomaterials in mice using laser-induced breakdown spectroscopy. J Adv Res 2020; 24:353-361. [PMID: 32489680 PMCID: PMC7256211 DOI: 10.1016/j.jare.2020.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/24/2020] [Accepted: 05/01/2020] [Indexed: 11/26/2022] Open
Abstract
Inorganic or inorganic-organic hybrid nanomaterials have great potential for applications in the biomedical fields. Biological half-life is an essential pharmacokinetic parameter for these materials to function in vivo. Compared to inductively coupled plasma mass spectrometry (ICP-MS), which is the gold standard, laser-induced breakdown spectroscopy (LIBS) is a faster and more efficient elemental detection method. We investigated an efficient way to quantify the metabolic rate using LIBS. Nanoparticle platforms, such as manganese dioxide-bovine serum albumin (MnO2-BSA) or boehmite-bovine serum albumin (AlO(OH)-BSA) were injected into mice through intravenous administration for LIBS spectrum acquisition. First, the spectral background was corrected using the polynomial fitting method; The spectral interference was eliminated by Lorentz fitting for each LIBS spectrum simultaneously. The support vector regression (SVR) was then used for LIBS quantitative analyses. Finally, the LIBS results were compared with the ICP-MS ones. The half-lives of MnO2-BSA calculated by LIBS and ICP-MS were 2.49 and 2.42 h, respectively. For AlO(OH)-BSA, the half-lives detected by LIBS and ICP-MS were 3.46 and 3.57 h, respectively. The relative error of LIBS is within 5% compared to ICP-MS. The results demonstrate that LIBS is a valuable tool for quantifying the metabolic rates with a high degree of accuracy.
Collapse
Affiliation(s)
- Yanwu Chu
- Wuhan National Laboratory for Optoelectronics (WNLO), Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Zhanjie Zhang
- Cancer Center, Union Hospital, TongjiMedical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Qianyuan He
- Cancer Center, Union Hospital, TongjiMedical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Feng Chen
- Wuhan National Laboratory for Optoelectronics (WNLO), Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Ziqian Sheng
- Wuhan National Laboratory for Optoelectronics (WNLO), Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Deng Zhang
- Wuhan National Laboratory for Optoelectronics (WNLO), Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Honglin Jin
- Cancer Center, Union Hospital, TongjiMedical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Fagang Jiang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lianbo Guo
- Wuhan National Laboratory for Optoelectronics (WNLO), Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|
17
|
Dogra P, Butner JD, Nizzero S, Ruiz Ramírez J, Noureddine A, Peláez MJ, Elganainy D, Yang Z, Le AD, Goel S, Leong HS, Koay EJ, Brinker CJ, Cristini V, Wang Z. Image-guided mathematical modeling for pharmacological evaluation of nanomaterials and monoclonal antibodies. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1628. [PMID: 32314552 PMCID: PMC7507140 DOI: 10.1002/wnan.1628] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/06/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022]
Abstract
While plasma concentration kinetics has traditionally been the predictor of drug pharmacological effects, it can occasionally fail to represent kinetics at the site of action, particularly for solid tumors. This is especially true in the case of delivery of therapeutic macromolecules (drug-loaded nanomaterials or monoclonal antibodies), which can experience challenges to effective delivery due to particle size-dependent diffusion barriers at the target site. As a result, disparity between therapeutic plasma kinetics and kinetics at the site of action may exist, highlighting the importance of target site concentration kinetics in determining the pharmacodynamic effects of macromolecular therapeutic agents. Assessment of concentration kinetics at the target site has been facilitated by non-invasive in vivo imaging modalities. This allows for visualization and quantification of the whole-body disposition behavior of therapeutics that is essential for a comprehensive understanding of their pharmacokinetics and pharmacodynamics. Quantitative non-invasive imaging can also help guide the development and parameterization of mathematical models for descriptive and predictive purposes. Here, we present a review of the application of state-of-the-art imaging modalities for quantitative pharmacological evaluation of therapeutic nanoparticles and monoclonal antibodies, with a focus on their integration with mathematical models, and identify challenges and opportunities. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Achraf Noureddine
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - María J Peláez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA.,Applied Physics Graduate Program, Rice University, Houston, Texas, USA
| | - Dalia Elganainy
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhen Yang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Anh-Dung Le
- Nanoscience and Microsystems Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - Shreya Goel
- Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hon S Leong
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Eugene J Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - C Jeffrey Brinker
- Department of Chemical and Biological Engineering and UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
18
|
Iriarte-Mesa C, López YC, Matos-Peralta Y, de la Vega-Hernández K, Antuch M. Gold, Silver and Iron Oxide Nanoparticles: Synthesis and Bionanoconjugation Strategies Aimed at Electrochemical Applications. Top Curr Chem (Cham) 2020; 378:12. [PMID: 31907672 DOI: 10.1007/s41061-019-0275-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/13/2019] [Indexed: 12/22/2022]
Abstract
Nanomaterials have revolutionized the sensing and biosensing fields, with the development of more sensitive and selective devices for multiple applications. Gold, silver and iron oxide nanoparticles have played a particularly major role in this development. In this review, we provide a general overview of the synthesis and characteristics of gold, silver and iron oxide nanoparticles, along with the main strategies for their surface functionalization with ligands and biomolecules. Finally, different architectures suitable for electrochemical applications are reviewed, as well as their main fabrication procedures. We conclude with some considerations from the authors' perspective regarding the promising use of these materials and the challenges to be faced in the near future.
Collapse
Affiliation(s)
- Claudia Iriarte-Mesa
- Laboratorio de Química Bioinorgánica, Departamento de Química General e Inorgánica, Facultad de Química, Universidad de La Habana, Zapata y G, Vedado, Plaza de la Revolución, 10 400, La Habana, Cuba
| | - Yeisy C López
- Laboratorio de Química Bioinorgánica, Departamento de Química General e Inorgánica, Facultad de Química, Universidad de La Habana, Zapata y G, Vedado, Plaza de la Revolución, 10 400, La Habana, Cuba.,Instituto Politécnico Nacional, Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada, Calzada Legaria 694, Col. Irrigación, 11 500, Ciudad de México, Mexico
| | - Yasser Matos-Peralta
- Laboratorio de Química Bioinorgánica, Departamento de Química General e Inorgánica, Facultad de Química, Universidad de La Habana, Zapata y G, Vedado, Plaza de la Revolución, 10 400, La Habana, Cuba
| | | | - Manuel Antuch
- Unité de Chimie et Procédés, École Nationale Supérieure de Techniques Avancées (ENSTA), Institut Polytechnique de Paris, 828 Boulevard des Maréchaux, 91120, Palaiseau, France.
| |
Collapse
|
19
|
Man F, Gawne PJ, T M de Rosales R. Nuclear imaging of liposomal drug delivery systems: A critical review of radiolabelling methods and applications in nanomedicine. Adv Drug Deliv Rev 2019; 143:134-160. [PMID: 31170428 PMCID: PMC6866902 DOI: 10.1016/j.addr.2019.05.012] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/25/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022]
Abstract
The integration of nuclear imaging with nanomedicine is a powerful tool for efficient development and clinical translation of liposomal drug delivery systems. Furthermore, it may allow highly efficient imaging-guided personalised treatments. In this article, we critically review methods available for radiolabelling liposomes. We discuss the influence that the radiolabelling methods can have on their biodistribution and highlight the often-overlooked possibility of misinterpretation of results due to decomposition in vivo. We stress the need for knowing the biodistribution/pharmacokinetics of both the radiolabelled liposomal components and free radionuclides in order to confidently evaluate the images, as they often share excretion pathways with intact liposomes (e.g. phospholipids, metallic radionuclides) and even show significant tumour uptake by themselves (e.g. some radionuclides). Finally, we describe preclinical and clinical studies using radiolabelled liposomes and discuss their impact in supporting liposomal drug development and clinical translation in several diseases, including personalised nanomedicine approaches.
Collapse
Affiliation(s)
- Francis Man
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Peter J Gawne
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom; London Centre for Nanotechnology, King's College London, Strand Campus, London WC2R 2LS, United Kingdom.
| |
Collapse
|