1
|
Wang H, Pestre H, Tan EKN, Wedemann L, Schuhmacher JS, Schuhmacher M, Stellacci F. Facile lipid nanoparticle size engineering approach via controllable fusion induced by depletion forces. J Colloid Interface Sci 2025; 691:137334. [PMID: 40147373 DOI: 10.1016/j.jcis.2025.137334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Abstract
Lipid nanoparticles (LNPs) are among the most promising drug delivery carriers in research and development, with one major clinical application being messenger RNA (mRNA) vaccine. Current LNP production methods have the limit of generating low polydispersity index (PDI; PDI < 0.1) only for relatively small particles (<100 nm). It is known that larger LNPs have desirable properties, for example, particles with diameters in the 100 to 200 nm range have good immunogenicity. Yet, these larger particles' large PDI limits their clinical translation because of concerns about manufacturing reproducibility and possible side effects. We report here a facile approach to produce large and monodisperse (100-200 nm, PDI < 0.1) LNPs. The approach is based on adding 10 kDa polyethylene glycol (PEG) to a solution containing smaller LNPs. We show that PEG-induced depletion forces lead to the fusion of LNPs to form particles of approximately double the original size while keeping the same starting PDI. We discuss the fusion mechanism and show the parameters it depends on. In particular, we show that the fusion leads to a decrease in the fraction of empty LNPs. We show that the purification for PEG after fusion is simple and complete, thus, we believe that this is a method for the production of large LNP with low PDI that has a lot of potential to find industrial use.
Collapse
Affiliation(s)
- Heyun Wang
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Station 12, CH-1015 Lausanne, Switzerland
| | - Hugo Pestre
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Station 12, CH-1015 Lausanne, Switzerland
| | - Emie-Kim Ngo Tan
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Station 12, CH-1015 Lausanne, Switzerland
| | - Linda Wedemann
- Institute of Bioengineering (IBI), École Polytechnique Fédérale de Lausanne (EPFL), Station 12, CH-1015 Lausanne, Switzerland
| | - Jan S Schuhmacher
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Station 12, CH-1015 Lausanne, Switzerland
| | - Milena Schuhmacher
- Institute of Bioengineering (IBI), École Polytechnique Fédérale de Lausanne (EPFL), Station 12, CH-1015 Lausanne, Switzerland
| | - Francesco Stellacci
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Station 12, CH-1015 Lausanne, Switzerland; Institute of Bioengineering (IBI), École Polytechnique Fédérale de Lausanne (EPFL), Station 12, CH-1015 Lausanne, Switzerland; Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Station 12, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
2
|
Kim K. Hybrid Systems of Gels and Nanoparticles for Cancer Therapy: Advances in Multifunctional Therapeutic Platforms. Gels 2025; 11:170. [PMID: 40136875 PMCID: PMC11941994 DOI: 10.3390/gels11030170] [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: 02/06/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Cancer is a global health concern. Various therapeutic approaches, including chemotherapy, photodynamic therapy, and immunotherapy, have been developed for cancer treatment. Silica nanoparticles, quantum dots, and metal-organic framework (MOF)-based nanomedicines have gained interest in cancer therapy because of their selective accumulation in tumors via the enhanced permeability and retention (EPR) effect. However, bare nanoparticles face challenges including poor biocompatibility, low stability, limited drug-loading capacity, and rapid clearance by the reticuloendothelial system (RES). Gels with unique three-dimensional network structures formed through various interactions such as covalent and hydrogen bonds are emerging as promising materials for addressing these challenges. Gel hybridization enhances biocompatibility, facilitates controlled drug release, and confers cancer-targeting abilities to nanoparticles. This review discusses gel-nanoparticle hybrid systems for cancer treatment developed in the past five years and analyzes the roles of gels in these systems.
Collapse
Affiliation(s)
- Kibeom Kim
- Department of Chemistry and Life Science, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
3
|
Zhivkov AM, Hristova SH, Popov TT. Anticancer Nanoparticle Carriers of the Proapoptotic Protein Cytochrome c. Pharmaceutics 2025; 17:305. [PMID: 40142969 PMCID: PMC11945056 DOI: 10.3390/pharmaceutics17030305] [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: 12/19/2024] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
This review discusses the literature data on the synthesis, physicochemical properties, and cytotoxicity of composite nanoparticles bearing the mitochondrial protein cytochrome c (cytC), which can act as a proapoptotic mediator in addition to its main function as an electron carrier in the electron transport chain. The introduction of exogenous cytC via absorption of carrier particles, the phagocytosis of colloid particles of submicrometric size, or the receptor-mediated endocytosis of nanoparticles in cancer cells, initiates the process of apoptosis-a multistage cascade of biochemical reactions leading to complete destruction of the cells. CytC-carrier composite particles have the potential for use in the treatment of neoplasms with superficial localization: skin, mouth, stomach, colon, etc. This approach can solve the two main problems of anticancer therapy: selectivity and non-toxicity. Selectivity is based on the incapability of the normal cell to absorb (nano)particles, except for the cells of the immune system. The use of cytC as a protein that normally functions in mitochondria is harmless for the macroorganism. In this review, the factors limiting cytotoxicity and the ways to increase it are discussed from the point of view of the physicochemical properties of the cytC-carrier particles. The different techniques used for the preparation of cytC-bearing colloids and nanoparticles are discussed. Articles reporting the achievement of high cytotoxicity with each of the techniques are critically analyzed.
Collapse
Affiliation(s)
- Alexandar M. Zhivkov
- Scientific Research Center, “St. Kliment Ohridski” Sofia University, 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria
| | - Svetlana H. Hristova
- Department of Medical Physics and Biophysics, Medical Faculty, Medical University—Sofia, Zdrave Str. 2, 1431 Sofia, Bulgaria
- Faculty of Physics, Sofia University, 5 James Bourchier Blvd., 1164 Sofia, Bulgaria
| | - Trifon T. Popov
- Medical Faculty, Medical University—Sofia, Zdrave Str. 2, 1431 Sofia, Bulgaria
| |
Collapse
|
4
|
Liu Y, Zhu F, He J, Liang M. Ferritin versus Liposomes: A Comparative Analysis of Protein- and Lipid-Based Drug Delivery Systems. Bioconjug Chem 2025; 36:127-135. [PMID: 39927978 DOI: 10.1021/acs.bioconjchem.4c00576] [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: 02/11/2025]
Abstract
Drug delivery systems (DDSs) are crucial for the controlled release and targeted delivery of therapeutic agents, enhancing the stability and specificity of small molecules, nucleic acids, or peptides and addressing challenges such as drug instability and poor tissue targeting, particularly in oncology. Over the past few decades, liposomes have become one of the most widely used DDSs due to their unique physicochemical properties and biocompatibility. In the 1990s, liposomes were approved by the FDA as the first nanomedicine for disease treatment. Ferritin, a natural protein with a hollow nanocage structure, shares many similarities in architecture and functionality with liposomes. As an innovative DDS, ferritin offers distinct advantages including inherent tumor-targeting capabilities and exceptional biocompatibility. Liposomes and ferritin represent, respectively, established and emerging approaches in drug delivery, both excelling in key features like encapsulation efficiency and biocompatibility, which align with the standards for pharmaceutical carriers. While liposomal formulations have been clinically used, challenges such as precision targeting remain unresolved. In contrast, although ferritins hold considerable promise for drug delivery, they have not yet been implemented in clinical practice. In this review, we provide a comprehensive analysis of ferritins and liposomes as drug delivery vehicles, evaluating their drug-loading capacities, tumor-targeting capabilities, biocompatibility, and therapeutic potential. On the basis of a comparison of their intended applications and inherent limitations in the context of current treatment strategies, ferritin is expected to be an ideal delivery vehicle for tumor-targeted therapy and a strong candidate for clinical translation in the near future.
Collapse
Affiliation(s)
- Yang Liu
- Experimental Center of Advanced Materials, School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Feiyan Zhu
- Experimental Center of Advanced Materials, School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Jiuyang He
- Experimental Center of Advanced Materials, School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Minmin Liang
- Experimental Center of Advanced Materials, School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
5
|
Pytlíková S, Konefał R, Pola R, Braunová A, Lobaz V, Šlouf M, Beneš H, Starenko D, Běhalová K, Kovář M, Etrych T, Laga R, Pechar M. Dual Thermo- and pH-Responsive Polymer Nanoparticle Assemblies for Potential Stimuli-Controlled Drug Delivery. ACS APPLIED BIO MATERIALS 2025; 8:271-284. [PMID: 39661715 PMCID: PMC11752510 DOI: 10.1021/acsabm.4c01167] [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: 08/16/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
The development of stimuli-responsive drug delivery systems enables targeted delivery and environment-controlled drug release, thereby minimizing off-target effects and systemic toxicity. We prepared and studied tailor-made dual-responsive systems (thermo- and pH-) based on synthetic diblock copolymers consisting of a fully hydrophilic block of poly[N-(1,3-dihydroxypropyl)methacrylamide] (poly(DHPMA)) and a thermoresponsive block of poly[N-(2,2-dimethyl-1,3-dioxan-5-yl)methacrylamide] (poly(DHPMA-acetal)) as drug delivery and smart stimuli-responsive materials. The copolymers were designed for eventual medical application to be fully soluble in aqueous solutions at 25 °C. However, they form well-defined nanoparticles with hydrodynamic diameters of 50-800 nm when heated above the transition temperature of 27-31 °C. This temperature range is carefully tailored to align with the human body's physiological conditions. The formation of the nanoparticles and their subsequent decomposition was studied using dynamic light scattering (DLS), transmission electron microscopy (TEM), isothermal titration calorimetry (ITC), and nuclear magnetic resonance (NMR). 1H NMR studies confirmed that after approximately 20 h of incubation at pH 5, which closely mimics tumor microenvironment, approximately 40% of the acetal groups were hydrolyzed, and the thermoresponsive behavior of the copolymers was lost. This smart polymer response led to disintegration of the supramolecular structures, possibly releasing the therapeutic cargo. By tuning the transition temperature to the values relevant for medical applications, we ensure precise and effective drug release. In addition, our systems did not exhibit any cytotoxicity against any of the three cell lines. Our findings underscore the immense potential of these nanoparticles as eventual advanced drug delivery systems, especially for cancer therapy.
Collapse
Affiliation(s)
- Sára Pytlíková
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague 6, 162 00, Czech Republic
| | - Rafal Konefał
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague 6, 162 00, Czech Republic
| | - Robert Pola
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague 6, 162 00, Czech Republic
| | - Alena Braunová
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague 6, 162 00, Czech Republic
| | - Volodymyr Lobaz
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague 6, 162 00, Czech Republic
| | - Miroslav Šlouf
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague 6, 162 00, Czech Republic
| | - Hynek Beneš
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague 6, 162 00, Czech Republic
| | - Daniil Starenko
- Institute
of Microbiology, Czech Academy of Sciences, Vídeňská 1083, Prague 4, 142 00, Czech Republic
| | - Kateřina Běhalová
- Institute
of Microbiology, Czech Academy of Sciences, Vídeňská 1083, Prague 4, 142 00, Czech Republic
| | - Marek Kovář
- Institute
of Microbiology, Czech Academy of Sciences, Vídeňská 1083, Prague 4, 142 00, Czech Republic
| | - Tomáš Etrych
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague 6, 162 00, Czech Republic
| | - Richard Laga
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague 6, 162 00, Czech Republic
| | - Michal Pechar
- Institute
of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, Prague 6, 162 00, Czech Republic
| |
Collapse
|
6
|
Ciftci F, Özarslan AC, Kantarci İC, Yelkenci A, Tavukcuoglu O, Ghorbanpour M. Advances in Drug Targeting, Drug Delivery, and Nanotechnology Applications: Therapeutic Significance in Cancer Treatment. Pharmaceutics 2025; 17:121. [PMID: 39861768 PMCID: PMC11769154 DOI: 10.3390/pharmaceutics17010121] [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: 12/03/2024] [Revised: 01/01/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
In the 21st century, thanks to advances in biotechnology and developing pharmaceutical technology, significant progress is being made in effective drug design. Drug targeting aims to ensure that the drug acts only in the pathological area; it is defined as the ability to accumulate selectively and quantitatively in the target tissue or organ, regardless of the chemical structure of the active drug substance and the method of administration. With drug targeting, conventional, biotechnological and gene-derived drugs target the body's organs, tissues, and cells that can be selectively transported to specific regions. These systems serve as drug carriers and regulate the timing of release. Despite having many advantageous features, these systems have limitations in thoroughly treating complex diseases such as cancer. Therefore, combining these systems with nanoparticle technologies is imperative to treat cancer at both local and systemic levels effectively. The nanocarrier-based drug delivery method involves encapsulating target-specific drug molecules into polymeric or vesicular systems. Various drug delivery systems (DDS) were investigated and discussed in this review article. The first part discussed active and passive delivery systems, hydrogels, thermoplastics, microdevices and transdermal-based drug delivery systems. The second part discussed drug carrier systems in nanobiotechnology (carbon nanotubes, nanoparticles, coated, pegylated, solid lipid nanoparticles and smart polymeric nanogels). In the third part, drug targeting advantages were discussed, and finally, market research of commercial drugs used in cancer nanotechnological approaches was included.
Collapse
Affiliation(s)
- Fatih Ciftci
- Department of Biomedical Engineering, Faculty of Engineering, Fatih Sultan Mehmet Vakıf University, Istanbul 34015, Turkey
- Department of Technology Transfer Office, Fatih Sultan Mehmet Vakıf University, Istanbul 34015, Turkey
| | - Ali Can Özarslan
- Department of Metallurgical and Materials Engineering, Istanbul University-Cerrahpasa, Istanbul 34320, Turkey;
| | - İmran Cagri Kantarci
- Department of Bioengineering, Faculty of Chemistry-Metallurgy, Yildiz Technical University, Istanbul 34210, Turkey;
| | - Aslihan Yelkenci
- Department of Pediatric Dentistry, Faculty of Dentistry, University of Health Sciences, Istanbul 34668, Turkey;
| | - Ozlem Tavukcuoglu
- Department of Biochemistry, Faculty of Hamidiye Pharmacy, University of Health Sciences, Istanbul 34668, Turkey;
| | - Mansour Ghorbanpour
- Department of Medicinal Plants, Faculty of Agriculture and Natural Resources, Arak University, Arak 38156-8-8349, Iran;
| |
Collapse
|
7
|
Achimas-Cadariu P, Kubelac PM, Pasca A, Gata VA, Fetica B, Balacescu O, Fischer-Fodor E, Focsan M, Astilean S, Vlad CI. Intraoperative imaging of residual ovarian cancer after neoadjuvant chemotherapy using indocyanine green. Int J Gynecol Cancer 2025:ijgc-2024-005568. [PMID: 39496391 DOI: 10.1136/ijgc-2024-005568] [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: 11/06/2024] Open
Abstract
OBJECTIVES Interval debulking surgery has similar outcomes and less morbidity compared with primary debulking in advanced ovarian cancer. However, there is controversy regarding the selection of chemotherapy-resistant clones. Complete resection is an essential prerequisite, and near-infrared surgery combined with various techniques for highlighting malignant foci strives to achieve actual complete resection. This study investigated the role of indocyanine green (ICG) in identifying additional residual malignant foci during interval debulking of apparently intact peritoneum not deemed clinically suspicious under white light inspection. METHODS Patients diagnosed with stage III or IV high-grade serous ovarian carcinoma, older than 18 years of age, with satisfactory hepatic and renal functions who underwent neoadjuvant chemotherapy according to the institutional protocol and were scheduled to undergo interval debulking surgery between 2020 and 2022 were deemed suitable for inclusion after agreeing to the study protocol and acknowledging no contraindications for the administration of the ICG product. After laparotomy and white light inspection, using bolus administration of ICG, additional suspect peritoneal samples in near infrared (defined by clinical hyper- or hypointensity areas compared with surrounding ICG fluorescence using the Zeiss Opmi Pentero 800 surgical microscope, that were not deemed clinically suspicious under white light) were excised. Descriptive statistics were inferred and the chi-square test was used for the comparison of excised areas. The Kaplan-Meier method was deployed for computing the overall survival and progression-free survival of the cohort. All statistical analyses were performed using IBM SPSS Statistics software. RESULTS Fifteen patients with a median age of 56 years were included. Most cases (n=10, 66.7%) were International Federation of Gynecology and Obstetrics (FIGO) stage III, and all patients received four to seven cycles of neoadjuvant platinum chemotherapy, with 40% of regimens using bevacizumab. The mean interval between neoadjuvant treatment and surgery was 39 (median 42, range 20-78) days. A total of 39 suspect additional peritoneal samples were analyzed, with 41% confirming malignant foci. The positive predictive value (PPV) for malignant foci was 30% in ICG hyperintense areas and 46% in ICG hypointense areas. Germline BRCA1/2 mutant patients and using neoadjuvant bevacizumab led to a higher PPV for ICG hypointense areas (60% and 72.7%, respectively). Overall, the number of additionally resected pathologically confirmed malignant lesions through ICG fluorescence increased by 25%. CONCLUSIONS The use of ICG was associated with an increase in the resection of samples with residual malignant foci. Overall, hypointense areas had a higher positive PPV for malignant foci in comparison with hyperintense ICG areas (46% vs 30%), which could be interpreted in the context of dynamic changes in the tumor microenvironment or enhanced permeability and retention effect following neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Patriciu Achimas-Cadariu
- Surgical Oncology, Oncology Institute Prof Dr Ion Chiricuta, Cluj-Napoca, Romania
- Surgical Oncology and Gynecologic Oncology, Iuliu Hagieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Paul Milan Kubelac
- Medical Oncology, Oncology Institute Prof Dr Ion Chiricuta, Cluj-Napoca, Romania
| | - Andrei Pasca
- Surgical Oncology, Oncology Institute Prof Dr Ion Chiricuta, Cluj-Napoca, Romania
- Surgical Oncology and Gynecologic Oncology, Iuliu Hagieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Vlad Alexandru Gata
- Surgical Oncology, Oncology Institute Prof Dr Ion Chiricuta, Cluj-Napoca, Romania
- Surgical Oncology and Gynecologic Oncology, Iuliu Hagieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Bogdan Fetica
- Pathology, Oncology Institute Prof Dr Ion Chiricuta, Cluj-Napoca, Romania
| | - Ovidiu Balacescu
- Genetics, Genomics and Experimental Pathology, Oncology Institute Prof Dr Ion Chiricuta, Cluj-Napoca, Romania
| | - Eva Fischer-Fodor
- Research Department, Oncology Institute Prof Dr Ion Chiricuta, Cluj-Napoca, Romania
| | - Monica Focsan
- Physics, Babeş-Bolyai University, Cluj-Napoca, Cluj, Romania
| | - Simion Astilean
- Physics, Babeş-Bolyai University, Cluj-Napoca, Cluj, Romania
| | - Catalin Ioan Vlad
- Surgical Oncology, Oncology Institute Prof Dr Ion Chiricuta, Cluj-Napoca, Romania
- Surgical Oncology and Gynecologic Oncology, Iuliu Hagieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| |
Collapse
|
8
|
Jun M, Vijayan V, Shin S, Nam HY, Song D, Choi J, Vasvani S, Cho SK, Park IK, Seo J. A bleomycin-mimicking manganese-porphyrin-conjugated mitochondria-targeting peptoid for cancer therapy. Bioorg Med Chem 2025; 117:118023. [PMID: 39602865 DOI: 10.1016/j.bmc.2024.118023] [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: 08/13/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
Bleomycin (BLM) is a natural product with established anticancer activity, attributed to its ability to cleave intracellular DNA. BLM complexes with iron (BLM-Fe3+) exhibit peroxidase-like activity, generate reactive oxygen species (ROS), and cause DNA cleavage. Inspired by the mechanism of BLM, we synthesized a novel conjugate of manganese tetraphenylporphyrin (MnTPP) with a biomimetic peptoid (i.e., oligo-N-substituted glycines); this conjugate harnesses the oxidative capabilities of manganese porphyrins combined with the cell-penetrating ability of a previously reported mitochondria-targeting peptoid (MTP). UV-vis spectroscopy showed the formation of Mn(V)-oxo porphyrin, a potent oxidative species, in the presence of hydrogen peroxide, simulating metallobleomycin reactivity. Biological assays demonstrated that MnTPP-MTP significantly boosted ROS production and induced cytotoxicity toward cancer cells, while sparing normal fibroblasts. Tetramethylrhodamine ethyl ester (TMRE) assay revealed reversible, dose-dependent impairment of the mitochondrial membrane potential by MnTPP-MTP treatment. DNA cleavage assays showed that MnTPP-MTP, specifically in the presence of hydrogen peroxide, could elicit substantial DNA damage, in a similar way to BLM. In vivo studies using liposome-encapsulated MnTPP-MTP (lipo-peptoid) indicated superior tumor suppression, without systemic toxicity, when administered locally. Immunofluorescence staining for Ki67 and TUNEL confirmed reduced cell proliferation and increased apoptosis, respectively, validating the anticancer efficacy of lipo-peptoid. These results suggest that MnTPP-MTP, particularly in a liposomal formulation, is a promising new chemotherapeutic agent with robust oxidative mechanisms, poised for further development and application against diverse cancers.
Collapse
Affiliation(s)
- Minjae Jun
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Veena Vijayan
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 58128, Republic of Korea
| | - Seungheon Shin
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Ho Yeon Nam
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Dasom Song
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jieun Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Shyam Vasvani
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 58128, Republic of Korea
| | - Steve K Cho
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 58128, Republic of Korea.
| | - Jiwon Seo
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
9
|
Liu D, Lu N, Zang F, Lu M, Zhang J, Zhao Y, Wan H, Wang M, Li QQ, Wang F, Luo S, Ma M, Shi F, Wu H, Tu J, Zhang Y. Magnetic Resonance Imaging-Based Radiogenomic Analysis Reveals Genomic Determinants for Nanoparticle Delivery into Tumors. ACS NANO 2024; 18:34615-34629. [PMID: 39663893 DOI: 10.1021/acsnano.4c09387] [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: 12/13/2024]
Abstract
Even though the enhanced permeability and retention (EPR) effect is applicable for the passive targeting of solid tumors, many nanodrugs have failed to achieve meaningful clinical outcomes due to the heterogeneity of EPR effect. Therefore, understanding the mechanism of the EPR effect is crucial to overcome the obstacles nanomedicines face in clinical translation. The aim of this study was to establish a reliable method to increase awareness of the critical influencing factors of nanoparticle (NP) transport into tumors based on the EPR effect using a combined radiogenomics and clinical magnetic resonance imaging (MRI) technique and gene set pathway enrichment analysis. Employing poly(lactic-co-glycolic acid) (PLGA)-coated Fe3O4 NPs as the contrast agent, the monolayer and multilayer distribution of the NPs were observed and quantitatively analyzed by MRI, improving the accuracy of evaluating vascular permeability by MRI. By performing Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of genes and pathways, we identified a variety of genes affecting vascular permeability, such as Cldn1, Dlg2, Bves, Prkag3, Cldn10, and Cldn8, which are related to tight junctions and control the permeability of blood vessels in tumors. The method presented here provides an MRI-supported approach to increase the breadth of data collected from genetic screens, reveals genetic evidence of the presence of NPs in tumors and lays a foundation for clinical patient stratification and personalized treatment.
Collapse
Affiliation(s)
- Di Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Na Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fengchao Zang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, School of Medicine, Southeast University, Nanjing 210096, P. R. China
| | - Mingze Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Jingyue Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Ying Zhao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Hao Wan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Mengjun Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Qian-Qian Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Shouhua Luo
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Ming Ma
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fangfang Shi
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210096, P. R. China
| | - Haoan Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Jing Tu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Yu Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| |
Collapse
|
10
|
Ando T, Vu TN, Nishimura T, Takahashi R, Yusa SI. Synthesis and Characterization of Polyion Complex Micelles with Glycopolymer Shells for Drug Delivery Carriers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:26249-26258. [PMID: 39591594 DOI: 10.1021/acs.langmuir.4c03795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Double hydrophilic diblock copolymers (G20A100 and G100A98), composed of non-charged poly(glycosyloxyethyl methacrylate) (PGEMA, G) and cationic poly((3-acrylamidopropyl)trimethylammonium chloride), were synthesized via reversible addition-fragementation chain transfer (RAFT) radical polymerization. Likewise, diblock copolymers (G20S80 and G100S78), composed of PGEMA and anionic poly(2-acrylamido-2-methylpropanesulfonate) were synthesized via RAFT. The subscripts in these abbreviations indicate the degree of polymerization (DP) of each block. Polyion complex (PIC) aggregates (G20A100/G20S80 and G100A98/G100S78) were formed through electrostatic interactions by combining oppositely charged diblock copolymers with matched DPs for charge neutralization. The hydrodynamic radii of the G20A100/G20S80 and G100A98/G100S78 PIC aggregates were 77.4 and 26.2 nm, respectively, with zeta potentials close to 0 mV. The G20A100/G20S80 PIC micelles tend to form intermicellar aggregates, resulting in an increase in the particle size over time. In contrast, G100A98/G100S78 PIC micelles exhibited colloidal stability with a constant spherical core-shell shape, which was unaffected by time. The morphology and stability of the PIC aggregates depend upon the DP ratio of PGEMA and oppositely charged polyelectrolyte blocks. Both G20A100/G20S80 and G100A98/G100S78 PIC aggregates dissociated above 0.8 M NaCl due to the screening effect of NaCl.
Collapse
Affiliation(s)
- Tomoki Ando
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji, Hyogo 671-2280, Japan
| | - Thi Ngan Vu
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji, Hyogo 671-2280, Japan
| | - Tomoya Nishimura
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji, Hyogo 671-2280, Japan
| | - Rintaro Takahashi
- Department of Macromolecular Science, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Shin-Ichi Yusa
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji, Hyogo 671-2280, Japan
| |
Collapse
|
11
|
Kim SY, Kim J, Kim H, Chang YT, Kwon HY, Lee JL, Yoon YS, Kim CW, Hong SM, Shin JH, Hong SW, Hwang SW, Ye BD, Byeon JS, Yang SK, Son BH, Myung SJ. Fluorescence-guided tumor visualization of colorectal cancer using tumor-initiating probe yellow in preclinical models. Sci Rep 2024; 14:26946. [PMID: 39505985 PMCID: PMC11542034 DOI: 10.1038/s41598-024-76312-1] [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: 04/25/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
Fluorescence-guided surgery has emerged as an innovative technique with promising applications in the treatment of various tumors, including colon cancer. Tumor-initiating probe yellow (TiY) has been discovered for identifying tumorigenic cells by unbiased phenotypic screening with thousands of diversity-oriented fluorescence library (DOFL) compounds in a patient-derived lung cancer cell model. This study demonstrated the clinical feasibility of TiY for tumor-specific fluorescence imaging in the tissues of patients with colorectal cancer (CRC). To evaluate the efficacy of TiY in tumor imaging, surgical specimens were obtained, consisting of 36 tissues from 18 patients with CRC, for ex vivo molecular fluorescence imaging, histology, and immunohistochemistry. Orthotopic and chemically induced CRC mice models were administered TiY topically, and distinct tumor lesions were observed in 10 min by real-time fluorescence colonoscopy and ex vivo imaging. In a hepatic metastasis mouse model using splenic injection, TiY accumulation was detected in metastatic liver lesions through fluorescence imaging. Correlation analysis between TiY intensity and protein expression, assessed via immunohistochemistry and Western blotting, revealed a positive correlation between TiY and vimentin and Zeb1, which are known as epithelial-mesenchymal transition (EMT) markers of cancers. A comparative analysis of TiY with other FDA-approved fluorescence probes such as ICG revealed greater quantitative differences in TiY fluorescence intensity between tumor and normal tissues than those observed with ICG. Altogether, these results demonstrated that TiY has a strong potential for visualizing CRC by fluorescence imaging in various preclinical models, which can be further translated for clinical use such as fluorescence-guided surgery. Furthermore, our data indicate that TiY is preferentially uptaken by cells with EMT induction and progression, and overexpressing vimentin and Zeb1 in patients with CRC.
Collapse
Affiliation(s)
- Sun Young Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jinhyeon Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hajung Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Tae Chang
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Hwa-Young Kwon
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Jong Lyul Lee
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yong Sik Yoon
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chan Wook Kim
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Mo Hong
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin-Ho Shin
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung Wook Hong
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung Wook Hwang
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byong Duk Ye
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong-Sik Byeon
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byung Ho Son
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Seung-Jae Myung
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Edis Biotech, Songpa-gu, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Gugu Nkosi PW, Chandran R, Abrahamse H. Hypocrellin: A Natural Photosensitizer and Nano-Formulation for Enhanced Molecular Targeting of PDT of Melanoma. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1997. [PMID: 39568119 PMCID: PMC11579242 DOI: 10.1002/wnan.1997] [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: 04/11/2024] [Revised: 06/29/2024] [Accepted: 09/10/2024] [Indexed: 11/22/2024]
Abstract
Nano-formulation has generated attention in the battle against cancer, because of its great flexibility, reduced adverse side effects, and accuracy in delivering drugs to target tissues dependent on the size and surface characteristics of the disease. The field of photodynamic treatment has advanced significantly in the past years. Photodynamic techniques that use nano-formulations have surfaced to further the field of nanotechnology in medicine, especially in cancer treatment. The pharmaceutical industry is seeing a growing trend toward enhanced drug formulation using nano-formulations such as liposomes, polymeric nanoparticles, dendrimers, nano-emulsions, and micelles. Natural extracts have also shown adverse effects when employed as photosensitizers in cancer therapy because they are cytotoxic when activated by light. Still, natural photosensitizers are a big part of cancer treatment. However, some shortcomings can be minimized by combining nano-formulations with these natural photosensitizers. The synergistic improvement in medication delivery that maintains or increases the mechanism of cell death in malignant cells has also been demonstrated by the combination of photodynamic therapy with nano-formulations and natural photosensitizers. Lastly, this review assesses the feasibility and potential of a photodynamic therapy system based on nano-formulations and natural photosensitizers in clinical treatment applications and briefly discusses the removal of toxic compounds associated with nano-formulations within cells.
Collapse
Affiliation(s)
| | - Rahul Chandran
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgDoornfonteinSouth Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgDoornfonteinSouth Africa
| |
Collapse
|
13
|
Lorenc P, Sikorska A, Molenda S, Guzniczak N, Dams-Kozlowska H, Florczak A. Physiological and tumor-associated angiogenesis: Key factors and therapy targeting VEGF/VEGFR pathway. Biomed Pharmacother 2024; 180:117585. [PMID: 39442237 DOI: 10.1016/j.biopha.2024.117585] [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: 10/03/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Cancer remains one of the leading causes of death worldwide and poses a significant challenge to effective treatment due to its complexity. Angiogenesis, the formation of new blood vessels, is one of the cancer hallmarks and is a critical process in tumor growth and metastasis. The pivotal role of angiogenesis in cancer development has made antiangiogenic treatment a promising strategy for cancer therapy. To develop an effective therapy, it is essential to understand the basics of the physiological and tumor angiogenesis process. This review presents the primary factors related to physiological and tumor angiogenesis and the mechanisms of angiogenesis in tumors. We summarize potential molecular targets for cancer treatment by focusing on the vasculature, with the VEGF/VEGFR pathway being one of the most important and well-studied. Additionally, we present the advantages and limitations of currently used clinical protocols for cancer treatment targeting the VEGF/VEGFR pathway.
Collapse
Affiliation(s)
- Patryk Lorenc
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61‑866, Poland; Doctoral School, Poznan University of Medical Sciences, 70 Bukowska St, Poznan 60-812, Poland
| | - Agata Sikorska
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61‑866, Poland
| | - Sara Molenda
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61‑866, Poland; Doctoral School, Poznan University of Medical Sciences, 70 Bukowska St, Poznan 60-812, Poland
| | - Natalia Guzniczak
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland
| | - Hanna Dams-Kozlowska
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61‑866, Poland
| | - Anna Florczak
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61‑866, Poland.
| |
Collapse
|
14
|
Szupryczyński K, Czeleń P, Jeliński T, Szefler B. What is the Reason That the Pharmacological Future of Chemotherapeutics in the Treatment of Lung Cancer Could Be Most Closely Related to Nanostructures? Platinum Drugs in Therapy of Non-Small and Small Cell Lung Cancer and Their Unexpected, Possible Interactions. The Review. Int J Nanomedicine 2024; 19:9503-9547. [PMID: 39296940 PMCID: PMC11410046 DOI: 10.2147/ijn.s469217] [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: 04/26/2024] [Accepted: 07/19/2024] [Indexed: 09/21/2024] Open
Abstract
Over the course of several decades, anticancer treatment with chemotherapy drugs for lung cancer has not changed significantly. Unfortunately, this treatment prolongs the patient's life only by a few months, causing many side effects in the human body. It has also been proven that drugs such as Cisplatin, Carboplatin, Oxaliplatin and others can react with other substances containing an aromatic ring in which the nitrogen atom has a free electron group in its structure. Thus, such structures may have a competitive effect on the nucleobases of DNA. Therefore, scientists are looking not only for new drugs, but also for new alternative ways of delivering the drug to the cancer site. Nanotechnology seems to be a great hope in this matter. Creating a new nanomedicine would reduce the dose of the drug to an absolute minimum, and thus limit the toxic effect of the drug; it would allow for the exclusion of interactions with competitive compounds with a structure similar to nucleobases; it would also permit using the so-called targeted treatment and bypassing healthy cells; it would allow for the introduction of other treatment options, such as radiotherapy directly to the cancer site; and it would provide diagnostic possibilities. This article is a review that aims to systematize the knowledge regarding the anticancer treatment of lung cancer, but not only. It shows the clear possibility of interactions of chemotherapeutics with compounds competitive to the nitrogenous bases of DNA. It also shows the possibilities of using nanostructures as potential Platinum drug carriers, and proves that nanomedicine can easily become a new medicinal product in personalized medicine.
Collapse
Affiliation(s)
- Kamil Szupryczyński
- Doctoral School of Medical and Health Sciences, Faculty of Pharmacy, Collegium Medicum, Nicolaus, Copernicus University, Bydgoszcz, Poland
| | - Przemysław Czeleń
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Jeliński
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Beata Szefler
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
15
|
Lin Q, Li J, Abudousalamu Z, Sun Y, Xue M, Yao L, Chen M. Advancing Ovarian Cancer Therapeutics: The Role of Targeted Drug Delivery Systems. Int J Nanomedicine 2024; 19:9351-9370. [PMID: 39282574 PMCID: PMC11401532 DOI: 10.2147/ijn.s478313] [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: 05/15/2024] [Accepted: 08/06/2024] [Indexed: 09/19/2024] Open
Abstract
Ovarian cancer (OC) is the most lethal reproductive system cancer and a leading cause of cancer-related death. The high mortality rate and poor prognosis of OC are primarily due to its tendency for extensive abdominal metastasis, late diagnosis in advanced stages, an immunosuppressive tumor microenvironment, significant adverse reactions to first-line chemotherapy, and the development of chemoresistance. Current adjuvant chemotherapies face challenges such as poor targeting, low efficacy, and significant side effects. Targeted drug delivery systems (TDDSs) are designed to deliver drugs precisely to the tumor site to enhance efficacy and minimize side effects. This review highlights recent advancements in the use of TDDSs for OC therapies, including drug conjugate delivery systems, nanoparticle drug delivery systems, and hydrogel drug delivery systems. The focus is on employing TDDS to conduct direct, effective, and safer interventions in OC through methods such as targeted tumor recognition and controlled drug release, either independently or in combination. This review also discusses the prospects and challenges for further development of TDDSs. Undoubtedly, the use of TDDSs shows promise in the battle against OCs.
Collapse
Affiliation(s)
- Qianhan Lin
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jiajia Li
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, People's Republic of China
| | - Zulimire Abudousalamu
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yating Sun
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, People's Republic of China
| | - Mengyang Xue
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, People's Republic of China
| | - Liangqing Yao
- Department of Gynecologic Oncology, Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Mo Chen
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
16
|
Boase NRB, Gillies ER, Goh R, Kieltyka RE, Matson JB, Meng F, Sanyal A, Sedláček O. Stimuli-Responsive Polymers at the Interface with Biology. Biomacromolecules 2024; 25:5417-5436. [PMID: 39197109 DOI: 10.1021/acs.biomac.4c00690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
There has been growing interest in polymeric systems that break down or undergo property changes in response to stimuli. Such polymers can play important roles in biological systems, where they can be used to control the release of therapeutics, modulate imaging signals, actuate movement, or direct the growth of cells. In this Perspective, after discussing the most important stimuli relevant to biological applications, we will present a selection of recent exciting developments. The growing importance of stimuli-responsive polysaccharides will be discussed, followed by a variety of stimuli-responsive polymeric systems for the delivery of small molecule drugs and nucleic acids. Switchable polymers for the emerging area of therapeutic response measurement in theranostics will be described. Then, the diverse functions that can be achieved using hydrogels cross-linked covalently, as well as by various dynamic approaches will be presented. Finally, we will discuss some of the challenges and future perspectives for the field.
Collapse
Affiliation(s)
- Nathan R B Boase
- Centre for Materials Science and School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Elizabeth R Gillies
- Department of Chemistry; Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Rubayn Goh
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Singapore 138634, Singapore
| | - Roxanne E Kieltyka
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, PO Box 9502, Leiden 2300 RA, The Netherlands
| | - John B Matson
- Department of Chemistry and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Amitav Sanyal
- Department of Chemistry and Center for Life Sciences and Technologies, Bogazici University, Bebek, 34342 Istanbul, Türkiye
| | - Ondřej Sedláček
- Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, 128 00 Prague 2, Czech Republic
| |
Collapse
|
17
|
Sun X, Zhou X, Shi X, Abed OA, An X, Lei YL, Moon JJ. Strategies for the development of metalloimmunotherapies. Nat Biomed Eng 2024; 8:1073-1091. [PMID: 38914800 PMCID: PMC11410547 DOI: 10.1038/s41551-024-01221-7] [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: 10/21/2022] [Accepted: 03/30/2024] [Indexed: 06/26/2024]
Abstract
Metal ions play crucial roles in the regulation of immune pathways. In fact, metallodrugs have a long record of accomplishment as effective treatments for a wide range of diseases. Here we argue that the modulation of interactions of metal ions with molecules and cells involved in the immune system forms the basis of a new class of immunotherapies. By examining how metal ions modulate the innate and adaptive immune systems, as well as host-microbiota interactions, we discuss strategies for the development of such metalloimmunotherapies for the treatment of cancer and other immune-related diseases.
Collapse
Affiliation(s)
- Xiaoqi Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Editas Medicine, Cambridge, MA, USA.
| | - Xingwu Zhou
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Xiaoyue Shi
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Omar A Abed
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Xinran An
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Yu Leo Lei
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Wu X, Feng S, Chang TS, Zhang R, Jaiswal S, Choi EYK, Duan Y, Jiang H, Wang TD. Detection of Hepatocellular Carcinoma in an Orthotopic Patient-Derived Xenograft with an Epithelial Cell Adhesion Molecule-Specific Peptide. Cancers (Basel) 2024; 16:2818. [PMID: 39199591 PMCID: PMC11352241 DOI: 10.3390/cancers16162818] [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: 06/24/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
Hepatocellular carcinoma (HCC) has emerged as a major contributor to the worldwide cancer burden. Improved methods are needed for early cancer detection and image-guided surgery. Peptides have small dimensions that can overcome delivery challenges to achieve high tumor concentrations and deep penetration. We used phage display methods to biopan against the extra-cellular domain of the purified EpCAM protein, and used IRDye800 as a near-infrared (NIR) fluorophore. The 12-mer sequence HPDMFTRTHSHN was identified, and specific binding to EpCAM was validated with HCC cells in vitro. A binding affinity of kd = 67 nM and onset of k = 0.136 min-1 (7.35 min) were determined. Serum stability was measured with a half-life of T1/2 = 2.6 h. NIR fluorescence images showed peak uptake in vivo by human HCC patient-derived xenograft (PDX) tumors at 1.5 h post-injection. Also, the peptide was able to bind to foci of local and distant metastases in liver and lung. Peptide biodistribution showed high uptake in tumor versus other organs. No signs of acute toxicity were detected during animal necropsy. Immunofluorescence staining of human liver showed specific binding to HCC compared with cirrhosis, adenoma, and normal specimens.
Collapse
Affiliation(s)
- Xiaoli Wu
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA; (X.W.); (S.F.); (S.J.)
| | - Shuo Feng
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA; (X.W.); (S.F.); (S.J.)
| | - Tse-Shao Chang
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Ruoliu Zhang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Sangeeta Jaiswal
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA; (X.W.); (S.F.); (S.J.)
| | - Eun-Young K. Choi
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Yuting Duan
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; (Y.D.); (H.J.)
| | - Hui Jiang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; (Y.D.); (H.J.)
| | - Thomas D. Wang
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA; (X.W.); (S.F.); (S.J.)
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
19
|
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
|
20
|
Hadzima M, Faucher FF, Blažková K, Yim JJ, Guerra M, Chen S, Woods EC, Park KW, Šácha P, Šubr V, Kostka L, Etrych T, Majer P, Konvalinka J, Bogyo M. Polymer-Tethered Quenched Fluorescent Probes for Enhanced Imaging of Tumor-Associated Proteases. ACS Sens 2024; 9:3720-3729. [PMID: 38941307 PMCID: PMC11287742 DOI: 10.1021/acssensors.4c00912] [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: 04/18/2024] [Revised: 05/30/2024] [Accepted: 06/12/2024] [Indexed: 06/30/2024]
Abstract
Fluorescence-based contrast agents enable real-time detection of solid tumors and their neovasculature, making them ideal for use in image-guided surgery. Several agents have entered late-stage clinical trials or secured FDA approval, suggesting they are likely to become the standard of care in cancer surgeries. One of the key parameters to optimize in contrast agents is molecular size, which dictates much of the pharmacokinetic and pharmacodynamic properties of the agent. Here, we describe the development of a class of protease-activated quenched fluorescent probes in which a N-(2-hydroxypropyl)methacrylamide copolymer is used as the primary scaffold. This copolymer core provides a high degree of probe modularity to generate structures that cannot be achieved with small molecules and peptide probes. We used a previously validated cathepsin substrate and evaluated the effects of length and type of linker, as well as the positioning of the fluorophore/quencher pair on the polymer core. We found that the polymeric probes could be optimized to achieve increased overall signal and tumor-to-background ratios compared to the reference small molecule probe. Our results also revealed multiple structure-activity relationship trends that can be used to design and optimize future optical imaging probes. Furthermore, they confirm that a hydrophilic polymer is an ideal scaffold for use in optical imaging contrast probes, allowing a highly modular design that enables efficient optimization to maximize probe accumulation and overall biodistribution properties.
Collapse
Affiliation(s)
- Martin Hadzima
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, Flemingovo n. 2, Praha 6 16610, Czech Republic
- Department
of Organic Chemistry, Faculty of Science, Charles University, Albertov 6, Praha 2 12800, Czech Republic
| | - Franco F. Faucher
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Kristýna Blažková
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Joshua J. Yim
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Matteo Guerra
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Shiyu Chen
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Emily C. Woods
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Ki Wan Park
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Pavel Šácha
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, Flemingovo n. 2, Praha 6 16610, Czech Republic
| | - Vladimír Šubr
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
n. 2, Praha 6 16206, Czech Republic
| | - Libor Kostka
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
n. 2, Praha 6 16206, Czech Republic
| | - Tomáš Etrych
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
n. 2, Praha 6 16206, Czech Republic
| | - Pavel Majer
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, Flemingovo n. 2, Praha 6 16610, Czech Republic
| | - Jan Konvalinka
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, Flemingovo n. 2, Praha 6 16610, Czech Republic
| | - Matthew Bogyo
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| |
Collapse
|
21
|
Li C, Wang M, Li PF, Sheng J, Fu Q. Construction of Smart DNA-Based Drug Delivery Systems for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306257. [PMID: 38377302 DOI: 10.1002/smll.202306257] [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: 07/24/2023] [Revised: 02/10/2024] [Indexed: 02/22/2024]
Abstract
Due to the disadvantages of poor targeting, slow action, and low effectiveness of current commonly used cancer treatments, including surgery, chemotherapy, and radiotherapy, researchers have turned to DNA as a biomaterial for constructing drug delivery nanocarriers. DNA is favored for its biocompatibility and programmability. In order to overcome the limitations associated with traditional drug delivery systems (DDSs), researchers have developed smart-responsive DNA DDSs that can control drug release in response to specific physical or chemical stimuli at targeted sites. In this review, a summary of multiple targeted ligand structures is provided, various shapes of stable DNA nanomaterials, and different stimuli-responsive drug release strategies in DNA DDSs. Specifically, targeted cell recognition, in vivo stable transport, and controlled drug release of smart DDSs are focused. Finally, the further development prospects and challenges of clinical application of DNA nanomaterials in the field of smart drug delivery are discussed. The objective of this review is to enhance researchers' comprehension regarding the potential application of DNA nanomaterials in precision drug delivery, with the aim of expediting the clinical implementation of intelligent DDSs.
Collapse
Affiliation(s)
- Congcong Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Mengzhen Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Pei-Feng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Junyue Sheng
- Qingdao No.58 High School of Shandong Province, 20 Jiushui Road, Qingdao, 266100, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
22
|
Saharkhiz S, Nasri N, Naderi N, Dini G, Ghalehshahi SS, Firoozbakht F. Evaluating a targeted Palbociclib-Trastuzumab loaded smart niosome platform for treating HER2 positive breast cancer cells. Int J Pharm X 2024; 7:100237. [PMID: 38516198 PMCID: PMC10955288 DOI: 10.1016/j.ijpx.2024.100237] [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: 12/08/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
In this study, we present a targeted and pH-sensitive niosomal (pHSN) formulation, incorporating quantum dot (QD)-labeled Trastuzumab (Trz) molecules for the specific delivery of Palbociclib (Pal) to cells overexpressing human epidermal growth factor receptor 2 (HER2). FTIR analyses confirmed the successful preparation of the pHSNs and their bioconjugation. The labeled Trz-conjugated Pal-pHSNs (Trz-Pal-pHSNs) exhibited a size of approximately 170 nm, displaying a spherical shape with a neutral surface charge of -1.2 mV. Pal encapsulation reached ∼86%, and the release pattern followed a two-phase pH-dependent mechanism. MTT assessments demonstrated enhanced apoptosis induction, particularly in HER2-positive cells, by Trz-Pal-pHSNs. Fluorescence imaging further validated the internalization of particles into cells. In conclusion, Trz-Pal-pHSNs emerge as a promising platform for personalized medicine in the treatment of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Shaghayegh Saharkhiz
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Negar Nasri
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Nazanin Naderi
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran 19839-69411, Iran
| | - Ghasem Dini
- Department of Nanotechnology, Faculty of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| | - Saeid Shirzadi Ghalehshahi
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Fateme Firoozbakht
- Department of Nanotechnology, Faculty of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| |
Collapse
|
23
|
Hadzima M, Faucher F, Blažková K, Yim JJ, Guerra M, Chen S, Woods EC, Park KW, Šácha P, Šubr V, Kostka L, Etrych T, Majer P, Konvalinka J, Bogyo M. Polymer-tethered quenched fluorescent probes for enhanced imaging of tumor associated proteases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592849. [PMID: 38766164 PMCID: PMC11100723 DOI: 10.1101/2024.05.06.592849] [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
Fluorescence-based contrast agents enable real-time detection of solid tumors and their neovasculature, making them ideal for use in image-guided surgery. Several agents have entered late-stage clinical trials or secured FDA approval, suggesting they are likely to become standard of care in cancer surgeries. One of the key parameters to optimize in contrast agent is molecular size, which dictates much of the pharmacokinetic and pharmacodynamic properties of the agent. Here, we describe the development of a class of protease-activated quenched fluorescent probes in which a N-(2-hydroxypropyl)methacrylamide copolymer is used as the primary scaffold. This copolymer core provides a high degree of probe modularity to generate structures that cannot be achieved with small molecules and peptide probes. We used a previously validated cathepsin substrate and evaluated the effects of length and type of linker as well as positioning of the fluorophore/quencher pair on the polymer core. We found that the polymeric probes could be optimized to achieve increased over-all signal and tumor-to-background ratios compared to the reference small molecule probe. Our results also revealed multiple structure-activity relationship trends that can be used to design and optimize future optical imaging probes. Furthermore, they confirm that a hydrophilic polymer is an ideal scaffold for use in optical imaging contrast probes, allowing a highly modular design that enables efficient optimization to maximize probe accumulation and overall biodistribution properties.
Collapse
Affiliation(s)
- Martin Hadzima
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo n. 2, 16610, Praha 6, Czech Republic
- Department of Organic Chemistry, Faculty of Science, Charles University, Albertov 6, 12800, Praha 2, Czech Republic
| | - Franco Faucher
- Department of Chemistry, Stanford University, Stanford, California, 94305, United States
- Department of Pathology, School of Medicine, Stanford University, Stanford, California, 94305, United States
| | - Kristýna Blažková
- Department of Pathology, School of Medicine, Stanford University, Stanford, California, 94305, United States
| | - Joshua J. Yim
- Department of Pathology, School of Medicine, Stanford University, Stanford, California, 94305, United States
| | - Matteo Guerra
- Department of Pathology, School of Medicine, Stanford University, Stanford, California, 94305, United States
| | - Shiyu Chen
- Department of Pathology, School of Medicine, Stanford University, Stanford, California, 94305, United States
| | - Emily C. Woods
- Department of Pathology, School of Medicine, Stanford University, Stanford, California, 94305, United States
| | - Ki Wan Park
- Department of Pathology, School of Medicine, Stanford University, Stanford, California, 94305, United States
| | - Pavel Šácha
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo n. 2, 16610, Praha 6, Czech Republic
| | - Vladimír Šubr
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského n. 2, 16206, Praha 6, Czech Republic
| | - Libor Kostka
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského n. 2, 16206, Praha 6, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského n. 2, 16206, Praha 6, Czech Republic
| | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo n. 2, 16610, Praha 6, Czech Republic
| | - Jan Konvalinka
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo n. 2, 16610, Praha 6, Czech Republic
| | - Matthew Bogyo
- Department of Pathology, School of Medicine, Stanford University, Stanford, California, 94305, United States
| |
Collapse
|
24
|
Heaton AR, Lechuga LM, Tangsangasaksri M, Ludwig KD, Fain SB, Mecozzi S. A stable, highly concentrated fluorous nanoemulsion formulation for in vivo cancer imaging via 19F-MRI. NMR IN BIOMEDICINE 2024; 37:e5100. [PMID: 38230415 PMCID: PMC10987282 DOI: 10.1002/nbm.5100] [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: 12/08/2022] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/18/2024]
Abstract
Magnetic resonance imaging (MRI) is a routine diagnostic modality in oncology that produces excellent imaging resolution and tumor contrast without the use of ionizing radiation. However, improved contrast agents are still needed to further increase detection sensitivity and avoid toxicity/allergic reactions associated with paramagnetic metal contrast agents, which may be seen in a small percentage of the human population. Fluorine-19 (19F)-MRI is at the forefront of the developing MRI methodologies due to near-zero background signal, high natural abundance of 100%, and unambiguous signal specificity. In this study, we have developed a colloidal nanoemulsion (NE) formulation that can encapsulate high volumes of the fluorous MRI tracer, perfluoro-[15-crown-5]-ether (PFCE) (35% v/v). These nanoparticles exhibit long-term (at least 100 days) stability and high PFCE loading capacity in formulation with our semifluorinated triblock copolymer, M2F8H18. With sizes of approximately 200 nm, these NEs enable in vivo delivery and passive targeting to tumors. Our diagnostic formulation, M2F8H18/PFCE NE, yielded in vivo 19F-MR images with a high signal-to-noise ratio up to 100 in a tumor-bearing mouse model at clinically relevant scan times. M2F8H18/PFCE NE circulated stably in the vasculature, accumulated in high concentration of an estimated 4-9 × 1017 19F spins/voxel at the tumor site, and cleared from most organs over the span of 2 weeks. Uptake by the mononuclear phagocyte system to the liver and spleen was also observed, most likely due to particle size. These promising results suggest that M2F8H18/PFCE NE is a favorable 19F-MR diagnostic tracer for further development in oncological studies and potential clinical translation.
Collapse
Affiliation(s)
- Alexa R. Heaton
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Ave Madison WI 53706, USA
| | - Lawrence M. Lechuga
- Department of Medical Physics, University of Wisconsin-Madison, 1111 Highland Ave Madison WI 53705, USA
| | | | - Kai D. Ludwig
- Department of Medical Physics, University of Wisconsin-Madison, 1111 Highland Ave Madison WI 53705, USA
| | - Sean B. Fain
- Department of Medical Physics, University of Wisconsin-Madison, 1111 Highland Ave Madison WI 53705, USA
- Department of Radiology, University of Iowa, 200 Hawkins Drive Iowa City IA 52242, USA
| | - Sandro Mecozzi
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Ave Madison WI 53706, USA
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Ave Madison WI 53705, USA
| |
Collapse
|
25
|
Kashiwada A, Taoka N, Chijimi Y, Noguchi K, Shigematsu K, Miura M, Suzuki T. Weakly acidic pH-responsive liposomal content release induced by histidine-modified agents. Org Biomol Chem 2024; 22:2844-2850. [PMID: 38516851 DOI: 10.1039/d4ob00216d] [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: 03/23/2024]
Abstract
Internal stimuli-responsive controlled release from liposomal vesicles is an innovative approach for site-specific delivery of therapeutic drugs. In this study, to enhance the endosomal pH control of drug release from liposomes, a series of histidine-modified pH-sensitive Cn-His (n = 8, 12, 18) agents were designed and utilized as triggers for liposomal content release. The pH-dependent properties of Cn-His-incorporated liposomes were characterized using dynamic light scattering, ζ-potential, and fluorescence spectroscopy. The liposomes maintained a relatively uniform size across all pH conditions. However, the ζ-potential exhibited positive values at endosomal acidic pH levels and neutral or negative values at physiological pH levels. Furthermore, acidic pH-dependent release of both polar content (carboxyfluorescein) and nonpolar content (Nile red) was observed from the Cn-His-incorporated liposomes. Notably, with C12-His as the pH sensitizer, the pH dependence of liposomal content release was significantly evident. This work establishes endosomal pH-controllable liposome platforms, laying the groundwork for developing clinically applicable triggered release formulations.
Collapse
Affiliation(s)
- Ayumi Kashiwada
- Department of Applied Molecular Chemistry, College of Industrial Technology, Nihon University, 1-2-1, Izumi-cho, Narashino, Chiba, 275-8575, Japan.
- Department of Applied Molecular Chemistry, Graduate School of Industrial Technology, Nihon University, 1-2-1, Izumi-cho, Narashino, Chiba, 275-8575, Japan
| | - Narumi Taoka
- Department of Applied Molecular Chemistry, College of Industrial Technology, Nihon University, 1-2-1, Izumi-cho, Narashino, Chiba, 275-8575, Japan.
| | - Yukiha Chijimi
- Department of Applied Molecular Chemistry, College of Industrial Technology, Nihon University, 1-2-1, Izumi-cho, Narashino, Chiba, 275-8575, Japan.
| | - Keiko Noguchi
- Department of Applied Molecular Chemistry, Graduate School of Industrial Technology, Nihon University, 1-2-1, Izumi-cho, Narashino, Chiba, 275-8575, Japan
| | - Karin Shigematsu
- Laboratory of Molecular Chemistry, Faculty of Basic Pharmaceutical Sciences, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi, Chiba, 274-8555, Japan
| | - Motofumi Miura
- Laboratory of Molecular Chemistry, Faculty of Basic Pharmaceutical Sciences, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi, Chiba, 274-8555, Japan
| | - Toyofumi Suzuki
- Laboratory of Pharmaceutics, Faculty of Applied Pharmaceutical Sciences, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi, Chiba, 274-8555, Japan
| |
Collapse
|
26
|
Appiah E, Nakamura H, Assumang A, Etrych T, Haratake M. Chemical modification of bradykinin-polymer conjugates for optimum delivery of nanomedicines to tumors. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 57:102744. [PMID: 38460653 DOI: 10.1016/j.nano.2024.102744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 02/15/2024] [Accepted: 03/01/2024] [Indexed: 03/11/2024]
Abstract
We recently prepared pH-responsive HPMA copolymer conjugates of bradykinin (P-BK), which release BK in response to the acidic tumor microenvironment, and found that administration of P-BK increased the tumor accumulation and therapeutic efficacy of nanomedicine. Because the release of BK from P-BK determines its onset of action, P-BKs with different release rates were prepared, and their properties were evaluated. The release kinetics were significantly altered by substitution proximal to hydrazone bond, release constant of methyl-substituted P-BK (P-MeBK) was approximately 4- and 80-fold higher than that of cyclopropyl-substituted P-BK (P-CPBK) and phenyl-substituted P-BK (P-PhBK). None of the P-BKs were active, but the release of BK restored their BK-like activity. Pre-administration of the P-BKs increased the tumor accumulation of nanomedicine in C26 tumor-bearing mice by 2- and 1.4-fold for P-MeBK and P-PhBK at 3 and 6 h. Altogether, this study provides insights into the design of pH-responsive nanodrugs with the desired release properties to target acidic lesions such as cancer and inflammation.
Collapse
Affiliation(s)
- Enoch Appiah
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan
| | - Hideaki Nakamura
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan.
| | - Anthony Assumang
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky Sq. 2, 162 06 Prague 6, Czech Republic
| | - Mamoru Haratake
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan
| |
Collapse
|
27
|
Genedy HH, Humbert P, Laoulaou B, Le Moal B, Fusellier M, Passirani C, Le Visage C, Guicheux J, Lepeltier É, Clouet J. MicroRNA-targeting nanomedicines for the treatment of intervertebral disc degeneration. Adv Drug Deliv Rev 2024; 207:115214. [PMID: 38395361 DOI: 10.1016/j.addr.2024.115214] [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: 12/13/2023] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
Low back pain stands as a pervasive global health concern, afflicting almost 80% of adults at some point in their lives with nearly 40% attributable to intervertebral disc degeneration (IVDD). As only symptomatic relief can be offered to patients there is a dire need for innovative treatments.Given the accumulating evidence that multiple microRNAs (miRs) are dysregulated during IVDD, they could have a huge potential against this debilitating condition. The way miRs can profoundly modulate signaling pathways and influence several cellular processes at once is particularly exciting to tackle this multifaceted disorder. However, miR delivery encounters extracellular and intracellular biological barriers. A promising technology to address this challenge is the vectorization of miRs within nanoparticles, providing both protection and enhancing their uptake within the scarce target cells of the degenerated IVD. This comprehensive review presents the diverse spectrum of miRs' connection with IVDD and demonstrates their therapeutic potential when vectorized in nanomedicines.
Collapse
Affiliation(s)
- Hussein H Genedy
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Paul Humbert
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| | - Bilel Laoulaou
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Brian Le Moal
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Marion Fusellier
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Department of Diagnostic Imaging, CRIP, ONIRIS, College of Veterinary Medicine, Food Science and Engineering, Nantes F-44307, France
| | | | - Catherine Le Visage
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| | - Élise Lepeltier
- Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France; Institut Universitaire de France (IUF), France.
| | - Johann Clouet
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| |
Collapse
|
28
|
Nagainallur Ravichandran S, Das D, Dayananda EK, Dey A, Banerjee A, Sun-Zhang A, Zhang H, Sun XF, Pathak S. A Review on Emerging Techniques for Diagnosis of Colorectal Cancer. Cancer Invest 2024; 42:119-140. [PMID: 38404236 DOI: 10.1080/07357907.2024.2315443] [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: 06/11/2023] [Accepted: 02/02/2024] [Indexed: 02/27/2024]
Abstract
Common detection methods in practice for diagnosing colorectal cancer (CRC) are painful and invasive leading to less participation of individuals for CRC diagnosis. Whereas, improved or enhanced imaging systems and other minimally invasive techniques with shorter detection times deliver greater detail and less discomfort in individuals. Thus, this review is a summary of the diagnostic tests, ranging from the simple potential use in developing a flexible CRC treatment to the patient's potential benefits in receiving less invasive procedures and the advanced treatments that might provide a better assessment for the diagnosis of CRC and reduce the mortality related to CRC.
Collapse
Affiliation(s)
- Shruthi Nagainallur Ravichandran
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| | - Diptimayee Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| | - Erica Katriel Dayananda
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| | - Amit Dey
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| | - Alexander Sun-Zhang
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden
| | - Hong Zhang
- Faculty of Medicine and Health, School of Medical Sciences, Orebro University, Örebro, Sweden
| | - Xiao-Feng Sun
- Division of Oncology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, India
| |
Collapse
|
29
|
Li H, Huang H, Tan H, Jia Q, Song W, Zhang Q, Zhou B, Bai J. Key processes in tumor metastasis and therapeutic strategies with nanocarriers: a review. Mol Biol Rep 2024; 51:197. [PMID: 38270746 DOI: 10.1007/s11033-023-08910-7] [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: 10/05/2023] [Accepted: 11/14/2023] [Indexed: 01/26/2024]
Abstract
Cancer metastasis is the leading cause of cancer-related death. Metastasis occurs at all stages of tumor development, with unexplored changes occurring at the primary site and distant colonization sites. The growing understanding of the metastatic process of tumor cells has contributed to the emergence of better treatment options and strategies. This review summarizes a range of features related to tumor cell metastasis and nanobased drug delivery systems for inhibiting tumor metastasis. The mechanisms of tumor metastasis in the ideal order of metastatic progression were summarized. We focus on the prominent role of nanocarriers in the treatment of tumor metastasis, summarizing the latest applications of nanocarriers in combination with drugs to target important components and processes of tumor metastasis and providing ideas for more effective nanodrug delivery systems.
Collapse
Affiliation(s)
- Hongjie Li
- School of Clinical Medicine, Weifang Medical University, 261053, Weifang, China
| | - Haiqin Huang
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China
| | - Haining Tan
- National Glycoengineering Research Center, Shandong University, 250012, Jinan, China
| | - Qitao Jia
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China
| | - Weina Song
- Department of Pediatric Respiratory and Critical Care, Qilu Hospital of Shandong University Dezhou Hospital, 253000, Dezhou, China
| | - Qingdong Zhang
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China.
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, 261053, Weifang, China.
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, 261053, Weifang, China.
| |
Collapse
|
30
|
Kastner A, Mendrina T, Babu T, Karmakar S, Poetsch I, Berger W, Keppler BK, Gibson D, Heffeter P, Kowol CR. Stepwise optimization of tumor-targeted dual-action platinum(iv)-gemcitabine prodrugs. Inorg Chem Front 2024; 11:534-548. [PMID: 38235273 PMCID: PMC10790623 DOI: 10.1039/d3qi02032k] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/28/2023] [Indexed: 01/19/2024]
Abstract
While platinum-based chemotherapeutic agents have established themselves as indispensable components of anticancer therapy, they are accompanied by a variety of side effects and the rapid occurrence of drug resistance. A promising strategy to address these challenges is the use of platinum(iv) prodrugs, which remain inert until they reach the tumor tissue, thereby mitigating detrimental effects on healthy cells. Typically, platinum drugs are part of combination therapy settings. Consequently, a very elegant strategy is the development of platinum(iv) prodrugs bearing a second, clinically relevant therapeutic in axial position. In the present study, we focused on gemcitabine as an approved antimetabolite, which is highly synergistic with platinum drugs. In addition, to increase plasma half-life and facilitate tumor-specific accumulation, an albumin-binding maleimide moiety was attached. Our investigations revealed that maleimide-cisplatin(iv)-gemcitabine complexes cannot carry sufficient amounts of gemcitabine to induce a significant effect in vivo. Consequently, we designed a carboplatin(iv) analog, that can be applied at much higher doses. Remarkably, this novel analog demonstrated impressive in vivo results, characterized by significant improvements in overall survival. Notably, these encouraging results could also be transferred to an in vivo xenograft model with acquired gemcitabine resistance, indicating the high potential of this approach.
Collapse
Affiliation(s)
- Alexander Kastner
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- University of Vienna, Vienna Doctoral School in Chemistry (DoSChem) Waehringer Str. 42 1090 Vienna Austria
| | - Theresa Mendrina
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria
| | - Tomer Babu
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem 9112102 Jerusalem Israel
| | - Subhendu Karmakar
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem 9112102 Jerusalem Israel
| | - Isabella Poetsch
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria
| | - Walter Berger
- Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| | - Bernhard K Keppler
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem 9112102 Jerusalem Israel
| | - Petra Heffeter
- Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| | - Christian R Kowol
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| |
Collapse
|
31
|
Naghib SM, Mohammad-Jafari K. Microfluidics-mediated Liposomal Nanoparticles for Cancer Therapy: Recent Developments on Advanced Devices and Technologies. Curr Top Med Chem 2024; 24:1185-1211. [PMID: 38424436 DOI: 10.2174/0115680266286460240220073334] [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: 12/09/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 03/02/2024]
Abstract
Liposomes, spherical particles with phospholipid double layers, have been extensively studied over the years as a means of drug administration. Conventional manufacturing techniques like thin-film hydration and extrusion have limitations in controlling liposome size and distribution. Microfluidics enables superior tuning of parameters during the self-assembly of liposomes, producing uniform populations. This review summarizes microfluidic methods for engineering liposomes, including hydrodynamic flow focusing, jetting, micro mixing, and double emulsions. The precise control over size and lamellarity afforded by microfluidics has advantages for cancer therapy. Liposomes created through microfluidics and designed to encapsulate chemotherapy drugs have exhibited several advantageous properties in cancer treatment. They showcase enhanced permeability and retention effects, allowing them to accumulate specifically in tumor tissues passively. This passive targeting of tumors results in improved drug delivery and efficacy while reducing systemic toxicity. Promising results have been observed in pancreatic, lung, breast, and ovarian cancer models, making them a potential breakthrough in cancer therapy. Surface-modified liposomes, like antibodies or carbohydrates, also achieve active targeting. Overall, microfluidic fabrication improves reproducibility and scalability compared to traditional methods while maintaining drug loading and biological efficacy. Microfluidics-engineered liposomal formulations hold significant potential to overcome challenges in nanomedicine-based cancer treatment.
Collapse
Affiliation(s)
- Seyed Morteza Naghib
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology, P.O. Box 16846-13114, Tehran, Iran
| | - Kave Mohammad-Jafari
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology, P.O. Box 16846-13114, Tehran, Iran
| |
Collapse
|
32
|
Kubeil M, Suzuki Y, Casulli MA, Kamal R, Hashimoto T, Bachmann M, Hayashita T, Stephan H. Exploring the Potential of Nanogels: From Drug Carriers to Radiopharmaceutical Agents. Adv Healthc Mater 2024; 13:e2301404. [PMID: 37717209 PMCID: PMC11468994 DOI: 10.1002/adhm.202301404] [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: 05/03/2023] [Revised: 08/21/2023] [Indexed: 09/18/2023]
Abstract
Nanogels open up access to a wide range of applications and offer among others hopeful approaches for use in the field of biomedicine. This review provides a brief overview of current developments of nanogels in general, particularly in the fields of drug delivery, therapeutic applications, tissue engineering, and sensor systems. Specifically, cyclodextrin (CD)-based nanogels are important because they have exceptional complexation properties and are highly biocompatible. Nanogels as a whole and CD-based nanogels in particular can be customized in a wide range of sizes and equipped with a desired surface charge as well as containing additional molecules inside and outside, such as dyes, solubility-mediating groups or even biological vector molecules for pharmaceutical targeting. Currently, biological investigations are mainly carried out in vitro, but more and more in vivo applications are gaining importance. Modern molecular imaging methods are increasingly being used for the latter. Due to an extremely high sensitivity and the possibility of obtaining quantitative data on pharmacokinetic and pharmacodynamic properties, nuclear methods such as single photon emission computed tomography (SPECT) and positron emission tomography (PET) using radiolabeled compounds are particularly suitable here. The use of radiolabeled nanogels for imaging, but also for therapy, is being discussed.
Collapse
Affiliation(s)
- Manja Kubeil
- Helmholtz‐Zentrum Dresden‐RossendorfInstitute of Radiopharmaceutical Cancer Research Bautzner Landstraße 40001328DresdenGermany
| | - Yota Suzuki
- Graduate School of Science and EngineeringSaitama University255 Shimo‐OkuboSakura‐KuSaitama338‐8570Japan
- Faculty of Science & TechnologySophia University7‐1 Kioi‐cho, Chiyoda‐kuTokyo102‐8554Japan
| | | | - Rozy Kamal
- Department of Nuclear MedicineManipal College of Health ProfessionsManipal Academy of Higher EducationManipalKarnataka576104India
| | - Takeshi Hashimoto
- Faculty of Science & TechnologySophia University7‐1 Kioi‐cho, Chiyoda‐kuTokyo102‐8554Japan
| | - Michael Bachmann
- Helmholtz‐Zentrum Dresden‐RossendorfInstitute of Radiopharmaceutical Cancer Research Bautzner Landstraße 40001328DresdenGermany
| | - Takashi Hayashita
- Faculty of Science & TechnologySophia University7‐1 Kioi‐cho, Chiyoda‐kuTokyo102‐8554Japan
| | - Holger Stephan
- Helmholtz‐Zentrum Dresden‐RossendorfInstitute of Radiopharmaceutical Cancer Research Bautzner Landstraße 40001328DresdenGermany
| |
Collapse
|
33
|
Wang Y, Song Y, He Y, Wang Y, Maurer J, Kiessling F, Lammers T, Wang F, Shi Y. Direct immunoactivation by chemotherapeutic drugs in cancer treatment. ADVANCED THERAPEUTICS 2023; 6:2300209. [PMID: 38249990 PMCID: PMC7615547 DOI: 10.1002/adtp.202300209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Indexed: 01/23/2024]
Abstract
The immune system plays a crucial role in recognizing and eliminating pathogenic substances and malignant cells in the body. For cancer treatment, immunotherapy is becoming the standard treatment for many types of cancer and is often combined with chemotherapy. Although chemotherapeutic agents are often reported to have adverse effects, including immunosuppression, they can also play a positive role in immunotherapy by directly stimulating the immune system. This has been demonstrated in preclinical and clinical studies in the past decades. Chemotherapeutics can activate immune cells through different immune receptors and signaling pathways depending on their chemical structure and formulation. In this review, we summarize and discuss the direct immunoactivation effects of chemotherapeutics and possible mechanisms behind these effects. Finally, we prospect chemo-immunotherapeutic combinations for the more effective and safer treatment of cancer.
Collapse
Affiliation(s)
- Yurui Wang
- Department of Polymer Therapeutics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Yiran Song
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Yazhi He
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Yang Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Jochen Maurer
- Department of Gynecology and Obstetrics, Uniklinik RWTH Aachen, Aachen 52074, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Feng Wang
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, PR China
| | - Yang Shi
- Department of Polymer Therapeutics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| |
Collapse
|
34
|
Balıbey FB, Bahadori F, Ergin Kizilcay G, Tekin A, Kanimdan E, Kocyigit A. Optimization of PLGA-DSPE hybrid nano-micelles with enhanced hydrophobic capacity for curcumin delivery. Pharm Dev Technol 2023; 28:843-855. [PMID: 37773031 DOI: 10.1080/10837450.2023.2264964] [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: 06/23/2023] [Accepted: 09/26/2023] [Indexed: 09/30/2023]
Abstract
Poly (D, L Lactic-co-Glycolic acid) (PLGA) is an FDA-approved polymer. It is distinguished from other biocompatible polymers by its feasibility of production and safety for intravenous cancer tumor targeting. Curcumin (CUR) is a natural molecule with versatile bioactivities including inhibiting the nuclear Factor kappa B (Nf-kB) levels in cancer cells, increased by chemotherapy agents. Our group previously reported a successful decrease in the p65 (RelA) subunit of Nf-kB using 125 µg/ml CUR loaded into PLGA nano-micelles. However, this amount was insufficient to reduce all Nf-kB subunits. This study aimed to increase the hydrophobic capacity of PLGA toward CUR using 1,2-Distearoyl-sn-glycerol-3-phosphoethanolamine (DSPE), an FDA-approved phospholipid. PLGA-DSPE hybrid nano-micelles (HNM) were prepared using two different methods, oil-in-water (OiWa) and film preparation-rehydration (FiRe). The encapsulated CUR was successfully increased to 250 µg/ml using the FiRe method. Physicochemical characterization of CUR-loaded HNM was performed using DLS FT-IR, DSC, and HPLC. In HNM with a size of 156.6 nm, DSPE, incorporated with all functional groups of PLGA, and CUR was trapped in the core of this structure. The release profile of CUR was suitable for targeted cancer therapy and the Encapsulation Efficacy was 92%.
Collapse
Affiliation(s)
- Fatmanur Babalı Balıbey
- Department of Biotechnology, Institute of Health Sciences, Bezmialem Vakif University, Fatih, Istanbul, Turkey
- Department of Medical Biochemistry, Bezmialem Vakif University, Fatih, Istanbul, Turkey
| | - Fatemeh Bahadori
- Department of Pharmaceutical Biotechnology, BezmialemVakif University, Istanbul, Turkey
- Department of Analytical Chemistry, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | | | - Adem Tekin
- Informatics Institute, Istanbul Technical University, Maslak, Istanbul, Turkey
| | - Ebru Kanimdan
- Department of Medical Biochemistry, Bezmialem Vakif University, Fatih, Istanbul, Turkey
| | - Abdurrahim Kocyigit
- Department of Medical Biochemistry, Bezmialem Vakif University, Fatih, Istanbul, Turkey
| |
Collapse
|
35
|
Wang Z, Xiao M, Guo F, Yan Y, Tian H, Zhang Q, Ren S, Yang L. Biodegradable polyester-based nano drug delivery system in cancer chemotherapy: a review of recent progress (2021-2023). Front Bioeng Biotechnol 2023; 11:1295323. [PMID: 38026861 PMCID: PMC10647934 DOI: 10.3389/fbioe.2023.1295323] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Cancer presents a formidable threat to human health, with the majority of cases currently lacking a complete cure. Frequently, chemotherapy drugs are required to impede its progression. However, these drugs frequently suffer from drawbacks such as poor selectivity, limited water solubility, low bioavailability, and a propensity for causing organ toxicity. Consequently, a concerted effort has been made to seek improved drug delivery systems. Nano-drug delivery systems based on biodegradable polyesters have emerged as a subject of widespread interest in this pursuit. Extensive research has demonstrated their potential for offering high bioavailability, effective encapsulation, controlled release, and minimal toxicity. Notably, poly (ε-caprolactone) (PCL), poly (lactic-co-glycolic acid) (PLGA), and polylactic acid (PLA) have gained prominence as the most widely utilized options as carriers of the nano drug delivery system. This paper comprehensively reviews recent research on these materials as nano-carriers for delivering chemotherapeutic drugs, summarizing their latest advancements, acknowledging their limitations, and forecasting future research directions.
Collapse
Affiliation(s)
- Zongheng Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
- Liaoning Research Institute of Family Planning (The Reproductive Hospital of China Medical University), Shenyang, China
| | - Miaomiao Xiao
- Liaoning Research Institute of Family Planning (The Reproductive Hospital of China Medical University), Shenyang, China
- College of Kinesiology, Shenyang Sport University, Shenyang, China
| | - Fangliang Guo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yue Yan
- Department of Emergency, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hong Tian
- Department of Oncology, The 4th People’s Hospital of Shenyang, China Medical University, Shenyang, China
| | - Qianshi Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shuangyi Ren
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Liqun Yang
- Liaoning Research Institute of Family Planning (The Reproductive Hospital of China Medical University), Shenyang, China
- Research Center for Biomedical Materials, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
36
|
Mahara A, Shima K, Soni R, Onishi R, Hirano Y, Saito S, Yamaoka T. In vivo MR imaging for tumor-associated initial neovascularization by supramolecular contrast agents. Colloids Surf B Biointerfaces 2023; 230:113525. [PMID: 37634287 DOI: 10.1016/j.colsurfb.2023.113525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Microvascular imaging is required to understand tumor angiogenesis development; however, an appropriate whole-body imaging method has not yet been established. Here, we successfully developed a supramolecular magnetic resonance (MR) contrast agent for long-term whole-tissue observation in a single individual. Fluorescein- and Gd-chelate-conjugated polyethylene glycols (PEGs) were synthesized, and their structures were optimized. Spectroscopic and pharmacokinetic analyses suggested that the fluorescein-conjugated linear and 8-arm PEGs with a molecular weight of approximately 10 kDa were suitable to form a supramolecular structure to visualize the microvessel structure and blood circulation. Microvascular formation was evaluated in a glioma cell transplantation model, and neovascularization around the glioma tissue at 5 days was observed, with the contrast agent leaking out into the cancer tissue. In contrast, after 12 days, microvessel structures were formed inside the glioma tissue, but the agents did not leak out. These imaging data for the first time proved that the microvessels formed inside cancer tissues at the early stage are very leaky, but that they form continuous microvessels after 12 days.
Collapse
Affiliation(s)
- Atsushi Mahara
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shim-machi, Suita, Osaka 564-8565, Japan.
| | - Keigo Shima
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shim-machi, Suita, Osaka 564-8565, Japan; Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35 Yamatecho, Suita, Osaka 565-8680, Japan
| | - Raghav Soni
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shim-machi, Suita, Osaka 564-8565, Japan
| | - Ryutaro Onishi
- Department of Medical Physics and Engineering, Graduate School of Medicine, Osaka University, 1-7 Yamadaoka, Suita, Osaka 565-0871, Japan; Division of Clinical Radiology Service, Kyoto University Hospital, Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshiaki Hirano
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35 Yamatecho, Suita, Osaka 565-8680, Japan
| | - Shigeyoshi Saito
- Department of Medical Physics and Engineering, Graduate School of Medicine, Osaka University, 1-7 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Advanced Medical Technologies, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shim-machi, Suita, Osaka 564-8565, Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shim-machi, Suita, Osaka 564-8565, Japan
| |
Collapse
|
37
|
Danchuk O, Levchenko A, da Silva Mesquita R, Danchuk V, Cengiz S, Cengiz M, Grafov A. Meeting Contemporary Challenges: Development of Nanomaterials for Veterinary Medicine. Pharmaceutics 2023; 15:2326. [PMID: 37765294 PMCID: PMC10536669 DOI: 10.3390/pharmaceutics15092326] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/30/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
In recent decades, nanotechnology has been rapidly advancing in various fields of human activity, including veterinary medicine. The review presents up-to-date information on recent advancements in nanotechnology in the field and an overview of the types of nanoparticles used in veterinary medicine and animal husbandry, their characteristics, and their areas of application. Currently, a wide range of nanomaterials has been implemented into veterinary practice, including pharmaceuticals, diagnostic devices, feed additives, and vaccines. The application of nanoformulations gave rise to innovative strategies in the treatment of animal diseases. For example, antibiotics delivered on nanoplatforms demonstrated higher efficacy and lower toxicity and dosage requirements when compared to conventional pharmaceuticals, providing a possibility to solve antibiotic resistance issues. Nanoparticle-based drugs showed promising results in the treatment of animal parasitoses and neoplastic diseases. However, the latter area is currently more developed in human medicine. Owing to the size compatibility, nanomaterials have been applied as gene delivery vectors in veterinary gene therapy. Veterinary medicine is at the forefront of the development of innovative nanovaccines inducing both humoral and cellular immune responses. The paper provides a brief overview of current topics in nanomaterial safety, potential risks associated with the use of nanomaterials, and relevant regulatory aspects.
Collapse
Affiliation(s)
- Oleksii Danchuk
- Institute of Climate-Smart Agriculture, National Academy of Agrarian Sciences, 24 Mayatska Road, Khlibodarske Village, 67667 Odesa, Ukraine;
| | - Anna Levchenko
- Department of Microbiology, Faculty of Veterinary Medicine, Ataturk University, Yakutiye, Erzurum 25240, Turkey;
| | | | - Vyacheslav Danchuk
- Ukrainian Laboratory of Quality and Safety of Agricultural Products, Mashynobudivna Str. 7, Chabany Village, 08162 Kyiv, Ukraine;
| | - Seyda Cengiz
- Milas Faculty of Veterinary Medicine, Mugla Sitki Kocman University, Mugla 48000, Turkey; (S.C.); (M.C.)
| | - Mehmet Cengiz
- Milas Faculty of Veterinary Medicine, Mugla Sitki Kocman University, Mugla 48000, Turkey; (S.C.); (M.C.)
| | - Andriy Grafov
- Department of Chemistry, University of Helsinki, A.I. Virtasen Aukio 1 (PL 55), 00560 Helsinki, Finland
| |
Collapse
|
38
|
Bauer T, Alberg I, Zengerling LA, Besenius P, Koynov K, Slütter B, Zentel R, Que I, Zhang H, Barz M. Tuning the Cross-Linking Density and Cross-Linker in Core Cross-Linked Polymeric Micelles and Its Effects on the Particle Stability in Human Blood Plasma and Mice. Biomacromolecules 2023; 24:3545-3556. [PMID: 37449781 PMCID: PMC10428167 DOI: 10.1021/acs.biomac.3c00308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/27/2023] [Indexed: 07/18/2023]
Abstract
Core cross-linked polymeric micelles (CCPMs) are designed to improve the therapeutic profile of hydrophobic drugs, reduce or completely avoid protein corona formation, and offer prolonged circulation times, a prerequisite for passive or active targeting. In this study, we tuned the CCPM stability by using bifunctional or trifunctional cross-linkers and varying the cross-linkable polymer block length. For CCPMs, amphiphilic thiol-reactive polypept(o)ides of polysarcosine-block-poly(S-ethylsulfonyl-l-cysteine) [pSar-b-pCys(SO2Et)] were employed. While the pCys(SO2Et) chain lengths varied from Xn = 17 to 30, bivalent (derivatives of dihydrolipoic acid) and trivalent (sarcosine/cysteine pentapeptide) cross-linkers have been applied. Asymmetrical flow field-flow fraction (AF4) displayed the absence of aggregates in human plasma, yet for non-cross-linked PM and CCPMs cross-linked with dihydrolipoic acid at [pCys(SO2Et)]17, increasing the cross-linking density or the pCys(SO2Et) chain lengths led to stable CCPMs. Interestingly, circulation time and biodistribution in mice of non-cross-linked and bivalently cross-linked CCPMs are comparable, while the trivalent peptide cross-linkers enhance the circulation half-life from 11 to 19 h.
Collapse
Affiliation(s)
- Tobias
A. Bauer
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Irina Alberg
- Department
of Chemistry, Johannes Gutenberg University
Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Lydia A. Zengerling
- Department
of Chemistry, Johannes Gutenberg University
Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Pol Besenius
- Department
of Chemistry, Johannes Gutenberg University
Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Kaloian Koynov
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Bram Slütter
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Rudolf Zentel
- Department
of Chemistry, Johannes Gutenberg University
Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Ivo Que
- Translational
Nanobiomaterials and Imaging Group, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333
ZA Leiden, The Netherlands
| | - Heyang Zhang
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Matthias Barz
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
- Department
of Dermatology, University Medical Center
of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
39
|
Li Y, Feng M, Guo T, Wang Z, Zhao Y. Tailored Beta-Lapachone Nanomedicines for Cancer-Specific Therapy. Adv Healthc Mater 2023; 12:e2300349. [PMID: 36970948 DOI: 10.1002/adhm.202300349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/16/2023] [Indexed: 03/29/2023]
Abstract
Nanotechnology shows the power to improve efficacy and reduce the adverse effects of anticancer agents. As a quinone-containing compound, beta-lapachone (LAP) is widely employed for targeted anticancer therapy under hypoxia. The principal mechanism of LAP-mediated cytotoxicity is believed due to the continuous generation of reactive oxygen species with the aid of NAD(P)H: quinone oxidoreductase 1 (NQO1). The cancer selectivity of LAP relies on the difference between NQO1 expression in tumors and that in healthy organs. Despite this, the clinical translation of LAP faces the problem of narrow therapeutic window that is challenging for dose regimen design. Herein, the multifaceted anticancer mechanism of LAP is briefly introduced, the advance of nanocarriers for LAP delivery is reviewed, and the combinational delivery approaches to enhance LAP potency in recent years are summarized. The mechanisms by which nanosystems boost LAP efficacy, including tumor targeting, cellular uptake enhancement, controlled cargo release, enhanced Fenton or Fenton-like reaction, and multidrug synergism, are also presented. The problems of LAP anticancer nanomedicines and the prospective solutions are discussed. The current review may help to unlock the potential of cancer-specific LAP therapy and speed up its clinical translation.
Collapse
Affiliation(s)
- Yaru Li
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| | - Meiyu Feng
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| | - Tao Guo
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300120, China
| | - Zheng Wang
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| | - Yanjun Zhao
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| |
Collapse
|
40
|
Ramirez-Garcia PD, Veldhuis NA, Bunnett NW, Davis TP. Targeting endosomal receptors, a new direction for polymers in nanomedicine. J Mater Chem B 2023; 11:5390-5399. [PMID: 37219363 PMCID: PMC10641892 DOI: 10.1039/d3tb00156c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
In this perspective, we outline a new opportunity for exploiting nanoparticle delivery of antagonists to target G-protein coupled receptors localized in intracellular compartments. We discuss the specific example of antagonizing endosomal receptors involved in pain to develop long-lasting analgesics but also outline the broader application potential of this delivery approach. We discuss the materials used to target endosomal receptors and indicate the design requirements for future successful applications.
Collapse
Affiliation(s)
- Paulina D Ramirez-Garcia
- Dentistry Translational Research Center, New York University College of Dentistry, New York, 10010, USA.
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Nicholas A Veldhuis
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone School of Medicine, New York, NY 10010, USA
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
41
|
Nemeth T, Yoshizawa-Sugata N, Pallier A, Tajima Y, Ma Y, Tóth É, Masai H, Yamakoshi Y. Water-Soluble Gd(III)-Porphyrin Complexes Capable of Both Photosensitization and Relaxation Enhancement. CHEMICAL & BIOMEDICAL IMAGING 2023; 1:157-167. [PMID: 37235189 PMCID: PMC10207321 DOI: 10.1021/cbmi.3c00007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023]
Abstract
With the aim of developing more stable Gd(III)-porphyrin complexes, two types of ligands 1 and 2 with carboxylic acid anchors were synthesized. Due to the N-substituted pyridyl cation attached to the porphyrin core, these porphyrin ligands were highly water-soluble and formed the corresponding Gd(III) chelates, Gd-1 and Gd-2. Gd-1 was sufficiently stable in neutral buffer, presumably due to the preferred conformation of the carboxylate-terminated anchors connected to nitrogen in the meta position of the pyridyl group helping to stabilize Gd(III) complexation by the porphyrin center. 1H NMRD (nuclear magnetic relaxation dispersion) measurements on Gd-1 revealed high longitudinal water proton relaxivity (r1 = 21.2 mM-1 s-1 at 60 MHz and 25 °C), which originates from slow rotational motion resulting from aggregation in aqueous solution. Under visible light irradiation, Gd-1 showed extensive photoinduced DNA cleavage in line with efficient photoinduced singlet oxygen generation. Cell-based assays revealed no significant dark cytotoxicity of Gd-1, while it showed sufficient photocytotoxicity on cancer cell lines under visible light irradiation. These results indicate the potential of this Gd(III)-porphyrin complex (Gd-1) as a core for the development of bifunctional systems acting as an efficient photodynamic therapy photosensitizer (PDT-PS) with magnetic resonance imaging (MRI) detection capabilities.
Collapse
Affiliation(s)
- Tamas Nemeth
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 3, CH8093 Zürich, Switzerland
| | - Naoko Yoshizawa-Sugata
- Research
Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo 156-8506, Japan
| | - Agnes Pallier
- Centre
de Biophysique Moléculaire, CNRS UPR 4301, University of Orléans, Rue Charles Sadron, 45071 Cedex 2 Orléans, France
| | - Youichi Tajima
- Department
of Basic Medical Sciences, Tokyo Metropolitan
Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo 156-8506, Japan
| | - Yue Ma
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 3, CH8093 Zürich, Switzerland
| | - Éva Tóth
- Centre
de Biophysique Moléculaire, CNRS UPR 4301, University of Orléans, Rue Charles Sadron, 45071 Cedex 2 Orléans, France
| | - Hisao Masai
- Department
of Basic Medical Sciences, Tokyo Metropolitan
Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo 156-8506, Japan
| | - Yoko Yamakoshi
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 3, CH8093 Zürich, Switzerland
| |
Collapse
|
42
|
Bauer TA, Schramm J, Fenaroli F, Siemer S, Seidl CI, Rosenauer C, Bleul R, Stauber RH, Koynov K, Maskos M, Barz M. Complex Structures Made Simple - Continuous Flow Production of Core Cross-Linked Polymeric Micelles for Paclitaxel Pro-Drug-Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210704. [PMID: 36934295 DOI: 10.1002/adma.202210704] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/10/2023] [Indexed: 05/26/2023]
Abstract
Translating innovative nanomaterials to medical products requires efficient manufacturing techniques that enable large-scale high-throughput synthesis with high reproducibility. Drug carriers in medicine embrace a complex subset of tasks calling for multifunctionality. Here, the synthesisof pro-drug-loaded core cross-linked polymeric micelles (CCPMs) in a continuous flow processis reported, which combines the commonly separated steps of micelle formation, core cross-linking, functionalization, and purification into a single process. Redox-responsive CCPMs are formed from thiol-reactive polypept(o)ides of polysarcosine-block-poly(S-ethylsulfonyl-l-cysteine) and functional cross-linkers based on dihydrolipoic acid hydrazide for pH-dependent release of paclitaxel. The precisely controlled microfluidic process allows the production of spherical micelles (Dh = 35 nm) with low polydispersity values (PDI < 0.1) while avoiding toxic organic solvents and additives with unfavorable safety profiles. Self-assembly and cross-linking via slit interdigital micromixers produces 350-700 mg of CCPMs/h per single system, while purification by online tangential flow filtration successfully removes impurities (unimer ≤ 0.5%). The formed paclitaxel-loaded CCPMs possess the desired pH-responsive release profile, display stable drug encapsulation, an improved toxicity profile compared to Abraxane (a trademark of Bristol-Myers Squibb), and therapeutic efficiency in the B16F1-xenotransplanted zebrafish model. The combination of reactive polymers, functional cross-linkers, and microfluidics enables the continuous-flow synthesis of therapeutically active CCPMs in a single process.
Collapse
Affiliation(s)
- Tobias A Bauer
- Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, Leiden, 2333CC, The Netherlands
| | - Jonas Schramm
- Fraunhofer Institute for Microengineering and Microsystems, Carl-Zeiss-Str. 18-20, 55129, Mainz, Germany
| | - Federico Fenaroli
- Department for Biosciences, University of Oslo, Blindernveien 31, 0371, Oslo, Norway
| | - Svenja Siemer
- Molecular and Cellular Oncology/Nanobiomedicine, ENT Department, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Christine I Seidl
- Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, Leiden, 2333CC, The Netherlands
| | - Christine Rosenauer
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Regina Bleul
- Fraunhofer Institute for Microengineering and Microsystems, Carl-Zeiss-Str. 18-20, 55129, Mainz, Germany
| | - Roland H Stauber
- Molecular and Cellular Oncology/Nanobiomedicine, ENT Department, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Michael Maskos
- Fraunhofer Institute for Microengineering and Microsystems, Carl-Zeiss-Str. 18-20, 55129, Mainz, Germany
| | - Matthias Barz
- Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, Leiden, 2333CC, The Netherlands
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| |
Collapse
|
43
|
Cai M, Yao Y, Yin D, Zhu R, Fu T, Kong J, Wang K, Liu J, Yao A, Ruan Y, Shi W, Zhu Q, Ni J, Yin X. Enhanced lysosomal escape of cell penetrating peptide-functionalized metal-organic frameworks for co-delivery of survivin siRNA and oridonin. J Colloid Interface Sci 2023; 646:370-380. [PMID: 37207419 DOI: 10.1016/j.jcis.2023.04.126] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/21/2023]
Abstract
In recent years, small interfering RNA (siRNA) has been widely used in the treatment of human diseases, especially tumors, and has shown great appeal. However, the clinical application of siRNA faces several challenges. Insufficient efficacy, poor bioavailability, poor stability, and lack of responsiveness to a single therapy are the main problems affecting tumor therapy. Here, we designed a cell-penetrating peptide (CPP)-modified metal organic framework nanoplatform (named PEG-CPP33@ORI@survivin siRNA@ZIF-90, PEG-CPP33@NPs) for targeted co-delivery of oridonin (ORI), a natural anti-tumor active ingredient) and survivin siRNA in vivo. This can improve the stability and bioavailability of siRNA and the efficacy of siRNA monotherapy. The high drug-loading capacity and pH-sensitive properties of zeolite imidazolides endowed the PEG-CPP33@NPs with lysosomal escape abilities. The Polyethylene glycol (PEG)-conjugated CPP (PEG-CPP33) coating significantly improved the uptake in the PEG-CPP33@NPs in vitro and in vivo. The results showed that the co-delivery of ORI and survivin siRNA greatly enhanced the anti-tumor effect of PEG-CPP33@NPs, demonstrating the synergistic effect between ORI and survivin siRNA. In summary, the novel targeted nanobiological platform loaded with ORI and survivin siRNA presented herein showed great advantages in cancer therapy, and provides an attractive strategy for the synergistic application of chemotherapy and gene therapy.
Collapse
Affiliation(s)
- Mengru Cai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yu Yao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Dongge Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Rongyue Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Tingting Fu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiahui Kong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Kaixin Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jing Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Aina Yao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yidan Ruan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wenjuan Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qian Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Ni
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xingbin Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
44
|
Qu H, Chen H, Cheng W, Wang Y, Xia Y, Zhang L, Ma B, Hu R, Xue X. A Supramolecular Assembly Strategy for Hydrophilic Drug Delivery towards Synergistic Cancer Treatment. Acta Biomater 2023; 164:407-421. [PMID: 37088157 DOI: 10.1016/j.actbio.2023.04.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/24/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
To improve the drug loading, tumor targeting, and delivery simplicity of hydrophilic drugs, we propose a supramolecular assembly strategy that potentially benefits a wide range of hydrophilic drug delivery. Firstly, we choose a hydrophilic drug (tirapazamine) as a model drug to directly co-assemble with chlorin e6 (Ce6) at different molar ratios, and systematically evaluate the resultant Ce6-tirapazamine nanoparticles (CT NPs) in aspects of size distribution, polydispersity, morphology, optical properties and molecular dynamics simulation. Based on the assembling facts between Ce6 and tirapazamine, we summarize a plausible rule of the supramolecular assembly for hydrophilic drugs. To validate our findings, more drugs with increasing hydrophilicity, such as temozolomide, gemcitabine hydrochloride and 5-azacytidine, successfully co-assemble with Ce6 into nanostructures by following similar assembling behaviors, demonstrating that our assembling rule may guide a wide range of hydrophilic drug delivery. Next, the combination of Ce6 and tirapazamine was chosen as the representative to investigate the anti-tumor activities of the supramolecular assemblies. CT NPs showed synergistic anti-tumor efficacy, increased tumor accumulation and significant tumor progression and metastasis inhibition in tumor-bearing mice. We anticipate that the supramolecular assembly mechanism will provide broad guidance for developing easy-to-make but functional nanomedicines. STATEMENT OF SIGNIFICANCE: Although thousands of nanomedicines have been developed, only a few have been approved for clinical use. The manufacturing complexity significantly hinders the "bench-to-bed" translation of nanomedicines. Hence, we need to rethink how to conduct research on translational nanomedicines by avoiding more and more complex chemistry and complicated nanostructures. Here, we summarize a plausible rule according to multiple supramolecular assembly pairs and propose a supramolecular assembly strategy that can improve the drug loading, tumor targeting, and manufacturing simplicity of nanomedicine for hydrophilic drugs. The supramolecular assembly strategy would guide a broader range of drug delivery to provide a new paradigm for developing easy-to-make but multifunctional nanoformulations for synergistic cancer treatment.
Collapse
Affiliation(s)
- Haijing Qu
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Han Chen
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei Cheng
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yanjun Wang
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China; Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Centre for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, China
| | - Yangyang Xia
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China; Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Centre for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, China
| | - Linghao Zhang
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Buyong Ma
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rong Hu
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Centre for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, China.
| | - Xiangdong Xue
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
45
|
Improved targeting delivery of WED-load immunoliposomes modified with SP-A mAb for the treatment of pulmonary fibrosis. Colloids Surf B Biointerfaces 2023; 224:113237. [PMID: 36871414 DOI: 10.1016/j.colsurfb.2023.113237] [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: 01/18/2023] [Revised: 02/20/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
The epithelial-mesenchymal transition (EMT) of type Ⅱ alveolar epithelial cells (AECS Ⅱ) induced by transforming growth factor (TGF-β1) is a primary pathogenesis of pulmonary fibrosis (PF). To augment the therapeutic potency of wedelolactone (WED) for PF, herein, pulmonary surfactant protein A (SP-A) specifically expressed on AECS Ⅱ was selected as the targeted receptor. Immunoliposomes modified with SP-A monoclonal antibody (SP-A mAb), novel anti-PF drug delivery systems, were developed and investigated in vivo and in vitro. In vivo fluorescence imaging technique was performed to evaluate the pulmonary-targeting effects of immunoliposomes. The result showed that immunoliposomes accumulated more in the lung, compared with non-modified nanoliposomes. Fluorescence detection methods and flow cytometry were used to investigate the function of SP-A mAb and the cellular uptake efficiency of WED-ILP in vitro. SP-A mAb enabled the immunoliposomes to specifically target the A549 cells and increased uptake more effectively. The mean fluorescence intensity (MFI) of cells treated with the targeted immunoliposomes was about 1.4-fold higher than that of cells treated with regular nanoliposomes. The cytotoxicity of nanoliposomes was assessed by the MTT assay, which demonstrated that blank nanoliposomes have no significant effect on A549 cell proliferation even at the SPC concentration of 1000 µg/mL. Additionally, in vitro pulmonary fibrosis model was established to further investigate the anti-pulmonary fibrosis effect of WED-ILP. WED-ILP significantly (**P < 0.01) inhibited the proliferation of A549 cells stimulated by TGF-β1 indicating that WED-ILP has great potential for the clinical treatment of PF.
Collapse
|
46
|
Stibbe JA, Hoogland P, Achterberg FB, Holman DR, Sojwal RS, Burggraaf J, Vahrmeijer AL, Nagengast WB, Rogalla S. Highlighting the Undetectable - Fluorescence Molecular Imaging in Gastrointestinal Endoscopy. Mol Imaging Biol 2023; 25:18-35. [PMID: 35764908 PMCID: PMC9971088 DOI: 10.1007/s11307-022-01741-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 11/27/2022]
Abstract
Flexible high-definition white-light endoscopy is the current gold standard in screening for cancer and its precursor lesions in the gastrointestinal tract. However, miss rates are high, especially in populations at high risk for developing gastrointestinal cancer (e.g., inflammatory bowel disease, Lynch syndrome, or Barrett's esophagus) where lesions tend to be flat and subtle. Fluorescence molecular endoscopy (FME) enables intraluminal visualization of (pre)malignant lesions based on specific biomolecular features rather than morphology by using fluorescently labeled molecular probes that bind to specific molecular targets. This strategy has the potential to serve as a valuable tool for the clinician to improve endoscopic lesion detection and real-time clinical decision-making. This narrative review presents an overview of recent advances in FME, focusing on probe development, techniques, and clinical evidence. Future perspectives will also be addressed, such as the use of FME in patient stratification for targeted therapies and potential alliances with artificial intelligence. KEY MESSAGES: • Fluorescence molecular endoscopy is a relatively new technology that enables safe and real-time endoscopic lesion visualization based on specific molecular features rather than on morphology, thereby adding a layer of information to endoscopy, like in PET-CT imaging. • Recently the transition from preclinical to clinical studies has been made, with promising results regarding enhancing detection of flat and subtle lesions in the colon and esophagus. However, clinical evidence needs to be strengthened by larger patient studies with stratified study designs. • In the future fluorescence molecular endoscopy could serve as a valuable tool in clinical workflows to improve detection in high-risk populations like patients with Barrett's esophagus, Lynch syndrome, and inflammatory bowel syndrome, where flat and subtle lesions tend to be malignant up to five times more often. • Fluorescence molecular endoscopy has the potential to assess therapy responsiveness in vivo for targeted therapies, thereby playing a role in personalizing medicine. • To further reduce high miss rates due to human and technical factors, joint application of artificial intelligence and fluorescence molecular endoscopy are likely to generate added value.
Collapse
Affiliation(s)
- Judith A Stibbe
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - Petra Hoogland
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Friso B Achterberg
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - Derek R Holman
- Department of Medicine, Division of Gastroenterology, Stanford University School of Medicine, Stanford, CA, USA
| | - Raoul S Sojwal
- Department of Medicine, Division of Gastroenterology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
- Centre for Human Drug Research, Leiden, The Netherlands
| | - Alexander L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - Wouter B Nagengast
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Stephan Rogalla
- Department of Medicine, Division of Gastroenterology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
47
|
Gautier L. Nanotechnology and cancer therapeutics: delivering on the hype? Biotechniques 2023; 74:63-67. [PMID: 36856138 DOI: 10.2144/btn-2022-0123] [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: 03/02/2023] Open
Abstract
Nanoparticle drug delivery systems are a promising development in cancer therapy for reducing toxicity and drug resistance, whilst improving precision targeting. Several types of nanotechnology are in development, with some receiving market approval and others proving difficult to translate to the bedside. [Formula: see text].
Collapse
|
48
|
Rosenkranz AA, Slastnikova TA, Durymanov MO, Georgiev GP, Sobolev AS. Exploiting active nuclear import for efficient delivery of Auger electron emitters into the cell nucleus. Int J Radiat Biol 2023; 99:28-38. [PMID: 32856963 DOI: 10.1080/09553002.2020.1815889] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND The most attractive features of Auger electrons (AEs) in cancer therapy are their extremely short range and sufficiently high linear energy transfer (LET) for a majority of them. The cytotoxic effects of AE emitters can be realized only in close vicinity to sensitive cellular targets and they are negligible if the emitters are located outside the cell. The nucleus is considered the compartment most sensitive to high LET particles. Therefore, the use of AE emitters could be most useful in specific recognition of a cancer cell and delivery of AE emitters into its nucleus. PURPOSE This review describes the studies aimed at developing effective anticancer agents for the delivery of AE emitters to the nuclei of target cancer cells. The use of peptide-based conjugates, nanoparticles, recombinant proteins, and other constructs for AE emitter targeted intranuclear delivery as well as their advantages and limitations are discussed. CONCLUSION Transport from the cytoplasm to the nucleus along with binding to the cancer cell is one of the key stages in the delivery of AE emitters; therefore, several constructs for exploitation of this transport have been developed. The transport is carried out through a nuclear pore complex (NPC) with the use of specific amino acid nuclear localization sequences (NLS) and carrier proteins named importins, which are located in the cytosol. Therefore, the effectiveness of NLS-containing delivery constructs designed to provide energy-dependent transport of AE emitter into the nuclei of cancer cells also depends on their efficient entry into the cytosol of the target cell.
Collapse
Affiliation(s)
- Andrey A Rosenkranz
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.,Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | | | | | - Alexander S Sobolev
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.,Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
49
|
Lin Z, Chu B, Qu Y, Wei X, Huang J, Wang F, Feng Y, Wang X, Luo H, Zhai X, Xu J, Liu X, Zhang L, Chen F, Wu Y, Zheng Y. Liposome-Encapsulated Melphalan Exhibits Potent Antimyeloma Activity and Reduced Toxicity. ACS OMEGA 2023; 8:1693-1701. [PMID: 36643473 PMCID: PMC9835516 DOI: 10.1021/acsomega.2c07555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023]
Abstract
Multiple myeloma (MM), a plasma cell cancer in bone marrow, remains an incurable disease. Melphalan, an alkylating agent, is a conventional anticancer drug that is still widely used for MM treatment in clinics. However, melphalan-induced organ toxicity and side effects are common. In this study, we loaded melphalan into a liposomal capsule and constituted liposomal melphalan (liposomal MEL). Liposomal MEL particles were approximately 120 nm in size and stable in vitro. The liposomal particles could be effectively taken up by MM cells. In vitro cytotoxicity assays using MM cell lines and primary MM cells showed that liposomal MEL exhibited similar anti-MM activity compared to an equivalent amount of free melphalan (free MEL) compound. In animal models, liposomal particles had bone marrow enrichment and prolonged half-life in vivo. Liposomal MEL exposure resulted in less liver and colon organ toxicity than exposure to an equivalent amount of free MEL-treated mice. Importantly, liposomal MEL had potent anti-MM activity in vivo in a human MM xenograft mouse model. Overall, our findings suggested that liposome-encapsulated melphalan was an effective drug modification of the melphalan compound and showed promise in MM treatment.
Collapse
Affiliation(s)
- Zhimei Lin
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
- Department
of Hematology, The Affiliated Hospital of
Chengdu University, Chengdu610081, P. R. China
| | - Bingyang Chu
- State
Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan610041, P. R. China
| | - Ying Qu
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
- State
Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan610041, P. R. China
| | - Xue Wei
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Jingcao Huang
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Fangfang Wang
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Yu Feng
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Xin Wang
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Hongmei Luo
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Xinyu Zhai
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Juan Xu
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Xiang Liu
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Li Zhang
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Fengjiao Chen
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Yu Wu
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| | - Yuhuan Zheng
- Department
of Hematology, West China Hospital, Sichuan
University, Chengdu610041, P. R. China
| |
Collapse
|
50
|
Role of Tunable Gold Nanostructures in Cancer Nanotheranostics: Implications on Synthesis, Toxicity, Clinical Applications and Their Associated Opportunities and Challenges. JOURNAL OF NANOTHERANOSTICS 2023. [DOI: 10.3390/jnt4010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The existing diagnosis and treatment modalities have major limitations related to their precision and capability to understand several stages of disease development. A superior therapeutic system consists of a multifunctional approach in early diagnosis of the disease with a simultaneous progressive cure, using a precise medical approach towards complex treatment. These challenges can be addressed via nanotheranostics and explore suitable approaches to improve health care. Nanotechnology in combination with theranostics as an unconventional platform paved the way for developing novel strategies and modalities leading to diagnosis and therapy for complex disease conditions, ranging from acute to chronic levels. Among the metal nanoparticles, gold nanoparticles are being widely used for theranostics due to their inherent non-toxic nature and plasmonic properties. The unique optical and chemical properties of plasmonic metal nanoparticles along with theranostics have led to a promising era of plausible early detection of disease conditions, and they enable real-time monitoring with enhanced non-invasive or minimally invasive imaging of several ailments. This review aims to highlight the improvement and advancement brought to nanotheranostics by gold nanoparticles in the past decade. The clinical use of the metal nanoparticles in nanotheranostics is explained, along with the future perspectives on addressing the key applications related to diagnostics and therapeutics, respectively. The scope of gold nanoparticles and their realistic potential to design a sophisticated theranostic system is discussed in detail, along with their implications in clinical advancements which are the needs of the hour. The review concluded with the challenges, opportunities, and implications on translational potential of using gold nanoparticles in nanotheranostics.
Collapse
|