1
|
Patle RY, Dongre RS. Recent advances in PAMAM mediated nano-vehicles for targeted drug delivery in Cancer Therapy. J Drug Target 2024:1-35. [PMID: 39530737 DOI: 10.1080/1061186x.2024.2428966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 10/02/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
3-D multi-faceted, nano-globular PAMAM dendritic skeleton is a highly significant polymer that offers applications in biomedical, industrial, environmental, and agricultural fields. This is mainly due to its enhanced properties, including adjustable surface functionalities, biocompatibility, non-toxicity, high uniformity, and reduced cytotoxicity, as well as its numerous internal cavities. This trait inspires further exploration and advancements in tailoring approaches. The implementation of deliberate strategic modifications in the morphological characteristics of PAMAM is crucial through chemical and biological interventions, in addition to its therapeutic advancements. Thus, the production of peripheral groups remains a prominent and highly advanced technique in molecular fabrication, aimed at boosting the potential of PAMAM conjugates. Currently, there exist numerous dendritic-hybrid materials, despite the widespread use of PAMAM-conjugated frameworks as drug delivery systems, which are well regarded for their efficacy in enhancing potency through the incorporation of surface functions. This paper provides a comprehensive review of recent progress in the design and assembly of various components of PAMAM-conjugates, focusing on their unique formulations. The review encompasses synthetic methodologies, a thorough evaluation of their applicability, and an analysis of their potential functions in the context of Drug Delivery Systems (DDS) in the current period.
Collapse
Affiliation(s)
- Ramkrishna Y Patle
- PGTD Chemistry, RTM Nagpur University, Nagpur-440033(M.S.), India
- Mahatma Gandhi College of Science, Gadchandur, Chandrapur-442908 (M.S.), India
| | | |
Collapse
|
2
|
Eftekhari Z, Zohrabi H, Oghalaie A, Ebrahimi T, Shariati FS, Behdani M, Kazemi-Lomedasht F. Advancements and challenges in mRNA and ribonucleoprotein-based therapies: From delivery systems to clinical applications. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102313. [PMID: 39281702 PMCID: PMC11402252 DOI: 10.1016/j.omtn.2024.102313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
The use of mRNA and ribonucleoproteins (RNPs) as therapeutic agents is a promising strategy for treating diseases such as cancer and infectious diseases. This review provides recent advancements and challenges in mRNA- and RNP-based therapies, focusing on delivery systems such as lipid nanoparticles (LNPs), which ensure efficient delivery to target cells. Strategies such as microfluidic devices are employed to prepare LNPs loaded with mRNA and RNPs, demonstrating effective genome editing and protein expression in vitro and in vivo. These applications extend to cancer treatment and infectious disease management, with promising results in genome editing for cancer therapy using LNPs encapsulating Cas9 mRNA and single-guide RNA. In addition, tissue-specific targeting strategies offer potential for improved therapeutic outcomes and reduced off-target effects. Despite progress, challenges such as optimizing delivery efficiency and targeting remain. Future research should enhance delivery efficiency, explore tissue-specific targeting, investigate combination therapies, and advance clinical translation. In conclusion, mRNA- and RNP-based therapies offer a promising avenue for treating various diseases and have the potential to revolutionize medicine, providing new hope for patients worldwide.
Collapse
Affiliation(s)
- Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Horieh Zohrabi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Tahereh Ebrahimi
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Fatemeh Sadat Shariati
- Department of Influenza and other Respiratory Viruses, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| |
Collapse
|
3
|
Chen P, Wang Z, Wang X, Gong J, Sheng J, Pan Y, Zhu D, Liu X. ROS-Responsive Ferrocenyl Amphiphilic PAMAM Dendrimers for On-Demand Delivery of siRNA Therapeutics to Cancer Cells. Pharmaceutics 2024; 16:936. [PMID: 39065632 PMCID: PMC11280363 DOI: 10.3390/pharmaceutics16070936] [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: 05/08/2024] [Revised: 06/26/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Small interfering RNA (siRNA) therapeutics, characterized by high specificity, potency, and durability, hold great promise in the treatment of cancer and other diseases. However, the clinic implementation of siRNA therapeutics critically depends on the safe and on-demand delivery of siRNA to the target cells. Here, we reported a family of ferrocenyl amphiphilic dendrimers (Fc-AmDs) for on-demand delivery of siRNA in response to the high ROS content in cancer cells. These dendrimers bear ROS-sensitive ferrocene moieties in the hydrophobic components and positively chargeable poly(amidoamine) dendrons as the hydrophilic entities, possessing favorable safety profiles and ROS responsive properties. One of these ferrocenyl amphiphilic dendrimers, Fc-C8-AmD 8A, outperforms in siRNA delivery, benefiting from its optimal balance of hydrophobicity and hydrophilicity. Its ROS feature facilitates specific and efficient disassembly of its complex with siRNA in ROS-rich cancer cells for effective siRNA delivery and gene silencing. Moreover, Fc-C8-AmD 8A also integrates the features and beneficial properties of both lipid and dendrimer vectors. Therefore, it represents a novel on-demand delivery system for cancer cell-specific siRNA delivery. This work opens new perspectives for designing self-assembly nanosystems for on-demand drug delivery.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dandan Zhu
- Center of Advanced Pharmaceuticals and Biomaterials, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; (P.C.); (Z.W.); (X.W.); (J.G.); (J.S.); (Y.P.)
| | - Xiaoxuan Liu
- Center of Advanced Pharmaceuticals and Biomaterials, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; (P.C.); (Z.W.); (X.W.); (J.G.); (J.S.); (Y.P.)
| |
Collapse
|
4
|
Lyu Z, Ralahy B, Perles-Barbacaru TA, Ding L, Jiang Y, Lian B, Roussel T, Liu X, Galanakou C, Laurini E, Tintaru A, Giorgio S, Pricl S, Liu X, Bernard M, Iovanna J, Viola A, Peng L. Self-assembling dendrimer nanosystems for specific fluorine magnetic resonance imaging and effective theranostic treatment of tumors. Proc Natl Acad Sci U S A 2024; 121:e2322403121. [PMID: 38865273 PMCID: PMC11194563 DOI: 10.1073/pnas.2322403121] [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: 12/20/2023] [Accepted: 05/13/2024] [Indexed: 06/14/2024] Open
Abstract
Fluorine magnetic resonance imaging (19F-MRI) is particularly promising for biomedical applications owing to the absence of fluorine in most biological systems. However, its use has been limited by the lack of safe and water-soluble imaging agents with high fluorine contents and suitable relaxation properties. We report innovative 19F-MRI agents based on supramolecular dendrimers self-assembled by an amphiphilic dendrimer composed of a hydrophobic alkyl chain and a hydrophilic dendron. Specifically, this amphiphilic dendrimer bears multiple negatively charged terminals with high fluorine content, which effectively prevented intra- and intermolecular aggregation of fluorinated entities via electrostatic repulsion. This permitted high fluorine nuclei mobility alongside good water solubility with favorable relaxation properties for use in 19F-MRI. Importantly, the self-assembling 19F-MRI agent was able to encapsulate the near-infrared fluorescence (NIRF) agent DiR and the anticancer drug paclitaxel for multimodal 19F-MRI and NIRF imaging of and theranostics for pancreatic cancer, a deadly disease for which there remains no adequate early detection method or efficacious treatment. The 19F-MRI and multimodal 19F-MRI and NIRF imaging studies on human pancreatic cancer xenografts in mice confirmed the capability of both imaging modalities to specifically image the tumors and demonstrated the efficacy of the theranostic agent in cancer treatment, largely outperforming the clinical anticancer drug paclitaxel. Consequently, these dendrimer nanosystems constitute promising 19F-MRI agents for effective cancer management. This study offers a broad avenue to the construction of 19F-MRI agents and theranostics, exploiting self-assembling supramolecular dendrimer chemistry.
Collapse
Affiliation(s)
- Zhenbin Lyu
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille13288, France
- Aix Marseille University, CNRS, Institut de Chimie Radicalaire, UMR 7273, Marseille13013, France
| | - Brigino Ralahy
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille13288, France
| | | | - Ling Ding
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille13288, France
- Aix Marseille University, CNRS, Centre de Résonance Magnétique Biologique et Médicale, UMR 7339, Marseille13385, France
| | - Yifan Jiang
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille13288, France
| | - Baoping Lian
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, People’s Republic of China
| | - Tom Roussel
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille13288, France
| | - Xi Liu
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille13288, France
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS, UMR 7258, Institut Paoli-Calmettes, Aix Marseille Université, Marseille13273, France
| | - Christina Galanakou
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille13288, France
| | - Erik Laurini
- Molecular Biology and Nanotechnology Laboratory, Department of Engineering and Architecture, University of Trieste, Trieste34127, Italy
| | - Aura Tintaru
- Aix Marseille University, CNRS, Institut de Chimie Radicalaire, UMR 7273, Marseille13013, France
| | - Suzanne Giorgio
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille13288, France
| | - Sabrina Pricl
- Molecular Biology and Nanotechnology Laboratory, Department of Engineering and Architecture, University of Trieste, Trieste34127, Italy
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz90-236, Poland
| | - Xiaoxuan Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, People’s Republic of China
| | - Monique Bernard
- Aix Marseille University, CNRS, Centre de Résonance Magnétique Biologique et Médicale, UMR 7339, Marseille13385, France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS, UMR 7258, Institut Paoli-Calmettes, Aix Marseille Université, Marseille13273, France
| | - Angèle Viola
- Aix Marseille University, CNRS, Centre de Résonance Magnétique Biologique et Médicale, UMR 7339, Marseille13385, France
| | - Ling Peng
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille13288, France
| |
Collapse
|
5
|
Yuan W, Shi X, Lee LTO. RNA therapeutics in targeting G protein-coupled receptors: Recent advances and challenges. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102195. [PMID: 38741614 PMCID: PMC11089380 DOI: 10.1016/j.omtn.2024.102195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
G protein-coupled receptors (GPCRs) are the major targets of existing drugs for a plethora of human diseases and dominate the pharmaceutical market. However, over 50% of the GPCRs remain undruggable. To pursue a breakthrough and overcome this situation, there is significant clinical research for developing RNA-based drugs specifically targeting GPCRs, but none has been approved so far. RNA therapeutics represent a unique and promising approach to selectively targeting previously undruggable targets, including undruggable GPCRs. However, the development of RNA therapeutics faces significant challenges in areas of RNA stability and efficient in vivo delivery. This review presents an overview of the advances in RNA therapeutics and the diverse types of nanoparticle RNA delivery systems. It also describes the potential applications of GPCR-targeted RNA drugs for various human diseases.
Collapse
Affiliation(s)
- Wanjun Yuan
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, People’s Republic of China
| | - Leo Tsz On Lee
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa 999078, Macau, China
| |
Collapse
|
6
|
El Riz A, Tchoumi Neree A, Mousavifar L, Roy R, Chorfi Y, Mateescu MA. Metallo-Glycodendrimeric Materials against Enterotoxigenic Escherichia coli. Microorganisms 2024; 12:966. [PMID: 38792795 PMCID: PMC11124148 DOI: 10.3390/microorganisms12050966] [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: 03/08/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Conjugation of carbohydrates to nanomaterials has been extensively studied and recognized as an alternative in the biomedical field. Dendrimers synthesized with mannose at the end group and with entrapped zero-valent copper/silver could be a potential candidate against bacterial proliferation. This study is aimed at investigating the bactericidal activity of metal-glycodendrimers. The Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) reaction was used to synthesize a new mannosylated dendrimer containing 12 mannopyranoside residues in the periphery. The enterotoxigenic Escherichia coli fimbriae 4 (ETEC:F4) viability, measured at 600 nm, showed the half-inhibitory concentration (IC50) of metal-free glycodendrimers (D), copper-loaded glycodendrimers (D:Cu) and silver-loaded glycodendrimers (D:Ag) closed to 4.5 × 101, 3.5 × 101 and to 1.0 × 10-2 µg/mL, respectively, and minimum inhibitory concentration (MIC) of D, D:Cu and D:Ag of 2.0, 1.5 and 1.0 × 10-4 µg/mL, respectively. The release of bacteria contents onto broth and the inhibition of ETEC:F4 biofilm formation increased with the number of metallo-glycodendrimer materials, with a special interest in silver-containing nanomaterial, which had the highest activity, suggesting that glycodendrimer-based materials interfered with bacteria-bacteria or bacteria-polystyrene interactions, with bacteria metabolism and can disrupt bacteria cell walls. Our findings identify metal-mannose-dendrimers as potent bactericidal agents and emphasize the effect of entrapped zero-valent metal against ETEC:F4.
Collapse
Affiliation(s)
- Aly El Riz
- Department of Chemistry, Université du Québec à Montréal, Succ. Centre-Ville, P.O. Box 8888, Montréal, QC H3C 3P8, Canada; (A.E.R.); (L.M.); (R.R.)
| | - Armelle Tchoumi Neree
- Department of Veterinary Biomedicine Sciences, Faculty of Veterinary Medicine, Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada; (A.T.N.); (Y.C.)
- Centre de recherche en infectiologie porcine et avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada
| | - Leila Mousavifar
- Department of Chemistry, Université du Québec à Montréal, Succ. Centre-Ville, P.O. Box 8888, Montréal, QC H3C 3P8, Canada; (A.E.R.); (L.M.); (R.R.)
| | - René Roy
- Department of Chemistry, Université du Québec à Montréal, Succ. Centre-Ville, P.O. Box 8888, Montréal, QC H3C 3P8, Canada; (A.E.R.); (L.M.); (R.R.)
| | - Younes Chorfi
- Department of Veterinary Biomedicine Sciences, Faculty of Veterinary Medicine, Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada; (A.T.N.); (Y.C.)
- Centre de recherche en infectiologie porcine et avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada
| | - Mircea Alexandru Mateescu
- Department of Chemistry, Université du Québec à Montréal, Succ. Centre-Ville, P.O. Box 8888, Montréal, QC H3C 3P8, Canada; (A.E.R.); (L.M.); (R.R.)
- Centre de recherche en infectiologie porcine et avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada
| |
Collapse
|
7
|
Gil-Cabrerizo P, Simon-Yarza T, Garbayo E, Blanco-Prieto MJ. Navigating the landscape of RNA delivery systems in cardiovascular disease therapeutics. Adv Drug Deliv Rev 2024; 208:115302. [PMID: 38574952 DOI: 10.1016/j.addr.2024.115302] [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: 11/28/2023] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
Cardiovascular diseases (CVDs) stand as the leading cause of death worldwide, posing a significant global health challenge. Consequently, the development of innovative therapeutic strategies to enhance CVDs treatment is imperative. RNA-based therapies, encompassing non-coding RNAs, mRNA, aptamers, and CRISPR/Cas9 technology, have emerged as promising tools for addressing CVDs. However, inherent challenges associated with RNA, such as poor cellular uptake, susceptibility to RNase degradation, and capture by the reticuloendothelial system, underscore the necessity of combining these therapies with effective drug delivery systems. Various non-viral delivery systems, including extracellular vesicles, lipid-based carriers, polymeric and inorganic nanoparticles, as well as hydrogels, have shown promise in enhancing the efficacy of RNA therapeutics. In this review, we offer an overview of the most relevant RNA-based therapeutic strategies explored for addressing CVDs and emphasize the pivotal role of delivery systems in augmenting their effectiveness. Additionally, we discuss the current status of these therapies and the challenges that hinder their clinical translation.
Collapse
Affiliation(s)
- Paula Gil-Cabrerizo
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Teresa Simon-Yarza
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science, INSERM U1148, X. Bichat Hospital, Paris 75018, France
| | - Elisa Garbayo
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain.
| | - María J Blanco-Prieto
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain.
| |
Collapse
|
8
|
Ding L, Lyu Z, Perles-Barbacaru TA, Huang AYT, Lian B, Jiang Y, Roussel T, Galanakou C, Giorgio S, Kao CL, Liu X, Iovanna J, Bernard M, Viola A, Peng L. Modular Self-Assembling Dendrimer Nanosystems for Magnetic Resonance and Multimodality Imaging of Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308262. [PMID: 38030568 DOI: 10.1002/adma.202308262] [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: 08/15/2023] [Revised: 11/26/2023] [Indexed: 12/01/2023]
Abstract
Bioimaging is a powerful tool for diagnosing tumors but remains limited in terms of sensitivity and specificity. Nanotechnology-based imaging probes able to accommodate abundant imaging units with different imaging modalities are particularly promising for overcoming these limitations. In addition, the nanosized imaging agents can specifically increase the contrast of tumors by exploiting the enhanced permeability and retention effect. A proof-of-concept study is performed on pancreatic cancer to demonstrate the use of modular amphiphilic dendrimer-based nanoprobes for magnetic resonance (MR) imaging (MRI) or MR/near-infrared fluorescence (NIRF) multimodality imaging. Specifically, the self-assembly of an amphiphilic dendrimer bearing multiple Gd3+ units at its terminals, generates a nanomicellar agent exhibiting favorable relaxivity for MRI with a good safety profile. MRI reveals an up to two-fold higher contrast enhancement in tumors than in normal muscle. Encapsulating the NIRF dye within the core of the nanoprobe yields an MR/NIRF bimodal imaging agent for tumor detection that is efficient both for MRI, at Gd3+ concentrations 1/10 the standard clinical dose, and for NIRF imaging, allowing over two-fold stronger fluorescence intensities. These self-assembling dendrimer nanosystems thus constitute effective probes for MRI and MR/NIRF multimodality imaging, offering a promising nanotechnology platform for elaborating multimodality imaging probes in biomedical applications.
Collapse
Affiliation(s)
- Ling Ding
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288, France
- Aix Marseille University, CNRS, Centre de Résonance Magnétique Biologique et Médicale (CRMBM), UMR 7339, Marseille, 13385, France
| | - Zhenbin Lyu
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288, France
| | - Teodora-Adriana Perles-Barbacaru
- Aix Marseille University, CNRS, Centre de Résonance Magnétique Biologique et Médicale (CRMBM), UMR 7339, Marseille, 13385, France
| | - Adela Ya-Ting Huang
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288, France
- Department of Medicinal and Applied Chemistry, Drug Development and Value Creation Research Center, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan
| | - Baoping Lian
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Yifan Jiang
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288, France
| | - Tom Roussel
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288, France
| | - Christina Galanakou
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288, France
| | - Suzanne Giorgio
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288, France
| | - Chai-Lin Kao
- Department of Medicinal and Applied Chemistry, Drug Development and Value Creation Research Center, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan
| | - Xiaoxuan Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS, UMR 7258, Institut Paoli-Calmettes, Aix Marseille Université, Marseille, 13273, France
| | - Monique Bernard
- Aix Marseille University, CNRS, Centre de Résonance Magnétique Biologique et Médicale (CRMBM), UMR 7339, Marseille, 13385, France
| | - Angèle Viola
- Aix Marseille University, CNRS, Centre de Résonance Magnétique Biologique et Médicale (CRMBM), UMR 7339, Marseille, 13385, France
| | - Ling Peng
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (UMR 7325), Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288, France
| |
Collapse
|
9
|
Malla R, Srilatha M, Farran B, Nagaraju GP. mRNA vaccines and their delivery strategies: A journey from infectious diseases to cancer. Mol Ther 2024; 32:13-31. [PMID: 37919901 PMCID: PMC10787123 DOI: 10.1016/j.ymthe.2023.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 11/04/2023] Open
Abstract
mRNA vaccines have evolved as promising cancer therapies. These vaccines can encode tumor-allied antigens, thus enabling personalized treatment approaches. They can also target cancer-specific mutations and overcome immune evasion mechanisms. They manipulate the body's cellular functions to produce antigens, elicit immune responses, and suppress tumors by overcoming limitations associated with specific histocompatibility leukocyte antigen molecules. However, successfully delivering mRNA into target cells destroys a crucial challenge. Viral and nonviral vectors (lipid nanoparticles and cationic liposomes) have shown great capacity in protecting mRNA from deterioration and assisting in cellular uptake. Cell-penetrating peptides, hydrogels, polymer-based nanoparticles, and dendrimers have been investigated to increase the delivery efficacy and immunogenicity of mRNA. This comprehensive review explores the landscape of mRNA vaccines and their delivery platforms for cancer, addressing design considerations, diverse delivery strategies, and recent advancements. Overall, this review contributes to the progress of mRNA vaccines as an innovative strategy for effective cancer treatment.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, AP, India
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati 517502, AP, India
| | - Batoul Farran
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA.
| |
Collapse
|
10
|
Cavazza A, Hendel A, Bak RO, Rio P, Güell M, Lainšček D, Arechavala-Gomeza V, Peng L, Hapil FZ, Harvey J, Ortega FG, Gonzalez-Martinez C, Lederer CW, Mikkelsen K, Gasiunas G, Kalter N, Gonçalves MA, Petersen J, Garanto A, Montoliu L, Maresca M, Seemann SE, Gorodkin J, Mazini L, Sanchez R, Rodriguez-Madoz JR, Maldonado-Pérez N, Laura T, Schmueck-Henneresse M, Maccalli C, Grünewald J, Carmona G, Kachamakova-Trojanowska N, Miccio A, Martin F, Turchiano G, Cathomen T, Luo Y, Tsai SQ, Benabdellah K. Progress and harmonization of gene editing to treat human diseases: Proceeding of COST Action CA21113 GenE-HumDi. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102066. [PMID: 38034032 PMCID: PMC10685310 DOI: 10.1016/j.omtn.2023.102066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
The European Cooperation in Science and Technology (COST) is an intergovernmental organization dedicated to funding and coordinating scientific and technological research in Europe, fostering collaboration among researchers and institutions across countries. Recently, COST Action funded the "Genome Editing to treat Human Diseases" (GenE-HumDi) network, uniting various stakeholders such as pharmaceutical companies, academic institutions, regulatory agencies, biotech firms, and patient advocacy groups. GenE-HumDi's primary objective is to expedite the application of genome editing for therapeutic purposes in treating human diseases. To achieve this goal, GenE-HumDi is organized in several working groups, each focusing on specific aspects. These groups aim to enhance genome editing technologies, assess delivery systems, address safety concerns, promote clinical translation, and develop regulatory guidelines. The network seeks to establish standard procedures and guidelines for these areas to standardize scientific practices and facilitate knowledge sharing. Furthermore, GenE-HumDi aims to communicate its findings to the public in accessible yet rigorous language, emphasizing genome editing's potential to revolutionize the treatment of many human diseases. The inaugural GenE-HumDi meeting, held in Granada, Spain, in March 2023, featured presentations from experts in the field, discussing recent breakthroughs in delivery methods, safety measures, clinical translation, and regulatory aspects related to gene editing.
Collapse
Affiliation(s)
- Alessia Cavazza
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, UK
| | - Ayal Hendel
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Rasmus O. Bak
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Paula Rio
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIEMAT/CIBERER), 28040 Madrid, Spain
- Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Marc Güell
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Integra Therapeutics S.L., Barcelona, Spain
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Virginia Arechavala-Gomeza
- Nucleic Acid Therapeutics for Rare Disorders (NAT-RD), Biobizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Ling Peng
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, Equipe Labellisée Ligue Contre le Cancer, 13288 Marseille, France
| | - Fatma Zehra Hapil
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Joshua Harvey
- Institute of Ophthalmology, University College London, London, UK
| | - Francisco G. Ortega
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Avenida de la Ilustración 114, 18016 Granada, Spain
- IBS Granada, Institute of Biomedical Research, Avenida de Madrid 15, 18012 Granada, Spain
| | - Coral Gonzalez-Martinez
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Avenida de la Ilustración 114, 18016 Granada, Spain
- IBS Granada, Institute of Biomedical Research, Avenida de Madrid 15, 18012 Granada, Spain
| | - Carsten W. Lederer
- Department of Molecular Genetics Thalassaemia, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Kasper Mikkelsen
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Nechama Kalter
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Manuel A.F.V. Gonçalves
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Julie Petersen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Alejandro Garanto
- Department of Pediatrics and Department of Human Genetics, Amalia Children’s Hospital, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lluis Montoliu
- Department of Molecular and Cellular Biology, National Centre for Biotechnology (CNB-CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain
| | - Marcello Maresca
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden
| | - Stefan E. Seemann
- Center for Non-coding RNA in Technology and Health, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jan Gorodkin
- Center for Non-coding RNA in Technology and Health, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Loubna Mazini
- Laboratory of Genetic Engineering, Technologic, Medical and Academic Park (TMAP), Marrakech, Morocco
| | - Rosario Sanchez
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of "Chemistry Applied to Biomedicine and the Environment," Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Universidad de Granada, Granada, Spain
| | - Juan R. Rodriguez-Madoz
- Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), Madrid, Spain
| | | | - Torella Laura
- DNA & RNA Medicine Division, Center for Applied Medical Research (CIMA) Universidad de Navarra, 31008 Pamplona, Spain
| | - Michael Schmueck-Henneresse
- Berlin Institute for Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany
| | - Cristina Maccalli
- Laboratory of Immune Biological Therapy, Research Branch, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Julian Grünewald
- Department of Medicine, Cardiology, Angiology, Pneumology, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, TranslaTUM, MIBE, Munich, Germany
- Center for Organoid Systems, Munich, Germany
| | - Gloria Carmona
- Red Andaluza de diseño y traslación de Terapias Avanzadas-RAdytTA, Fundación Pública Andaluza Progreso y Salud-FPS, Sevilla, España
| | | | - Annarita Miccio
- Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, Université de Paris Cité, INSERM UMR 1163, 75015 Paris, France
| | - Francisco Martin
- Bioquímica y Biología Molecular III e Immunology Department, Facultad de Medicina, Universidad de Granada and Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Av. de la Ilustración 114, 18016 Granada, Spain
| | - Giandomenico Turchiano
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, UK
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany
- Medical Faculty, University of Freiburg, 79106 Freiburg, Germany
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Shengdar Q. Tsai
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Karim Benabdellah
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Av. de la Ilustración 114, 18016 Granada, Spain
| | - on behalf of the COST Action CA21113
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, UK
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIEMAT/CIBERER), 28040 Madrid, Spain
- Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Integra Therapeutics S.L., Barcelona, Spain
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- Nucleic Acid Therapeutics for Rare Disorders (NAT-RD), Biobizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, Equipe Labellisée Ligue Contre le Cancer, 13288 Marseille, France
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
- Institute of Ophthalmology, University College London, London, UK
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Avenida de la Ilustración 114, 18016 Granada, Spain
- IBS Granada, Institute of Biomedical Research, Avenida de Madrid 15, 18012 Granada, Spain
- Department of Molecular Genetics Thalassaemia, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- CasZyme, 10224 Vilnius, Lithuania
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
- Department of Pediatrics and Department of Human Genetics, Amalia Children’s Hospital, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Molecular and Cellular Biology, National Centre for Biotechnology (CNB-CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain
- Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, Gothenburg, Sweden
- Center for Non-coding RNA in Technology and Health, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
- Laboratory of Genetic Engineering, Technologic, Medical and Academic Park (TMAP), Marrakech, Morocco
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of "Chemistry Applied to Biomedicine and the Environment," Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Universidad de Granada, Granada, Spain
- Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), Madrid, Spain
- DNA & RNA Medicine Division, Center for Applied Medical Research (CIMA) Universidad de Navarra, 31008 Pamplona, Spain
- Berlin Institute for Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany
- Laboratory of Immune Biological Therapy, Research Branch, Sidra Medicine, PO Box 26999, Doha, Qatar
- Department of Medicine, Cardiology, Angiology, Pneumology, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, TranslaTUM, MIBE, Munich, Germany
- Center for Organoid Systems, Munich, Germany
- Red Andaluza de diseño y traslación de Terapias Avanzadas-RAdytTA, Fundación Pública Andaluza Progreso y Salud-FPS, Sevilla, España
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
- Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, Université de Paris Cité, INSERM UMR 1163, 75015 Paris, France
- Bioquímica y Biología Molecular III e Immunology Department, Facultad de Medicina, Universidad de Granada and Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Av. de la Ilustración 114, 18016 Granada, Spain
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany
- Medical Faculty, University of Freiburg, 79106 Freiburg, Germany
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus N, Denmark
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), PTS, Av. de la Ilustración 114, 18016 Granada, Spain
| |
Collapse
|
11
|
Zhang P, Li Z, Cao W, Tang J, Xia Y, Peng L, Ma J. A PD-L1 Antibody-Conjugated PAMAM Dendrimer Nanosystem for Simultaneously Inhibiting Glycolysis and Promoting Immune Response in Fighting Breast Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2305215. [PMID: 37522451 DOI: 10.1002/adma.202305215] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/26/2023] [Indexed: 08/01/2023]
Abstract
Breast cancer is the most frequent malignancy affecting women, yet current therapeutic strategies remain ineffective for patients with late-stage or metastatic disease. Here an effective strategy is reported for treating metastatic breast cancer. Specifically, a self-assembling dendrimer nanosystem decorated with an antibody against programmed cell death ligand 1 (PD-L1) is established for delivering a small interfering RNA (siRNA) to target 3-phosphoinositide-dependent protein kinase-1 (PDK1), a kinase involved in cancer metabolism and metastasis. This nanosystem, named PPD, is designed to target PD-L1 for cancer-specific delivery of the siRNA to inhibit PDK1 and modulate cancer metabolism while promoting programmed cell death 1 (PD-1)/PD-L1 pathway-based immunotherapy. Indeed, PPD effectively generates simultaneous inhibition of PDK1-induced glycolysis and the PD-1/PD-L1 pathway-related immune response, leading to potent inhibition of tumor growth and metastasis without any notable toxicity in tumor-bearing mouse models. Collectively, these results highlight the potential use of PPD as an effective and safe tumor-targeting therapy for breast cancer. This study constitutes a successful proof of principle exploiting the intrinsic features of the tumor microenvironment and metabolism alongside a unique self-assembling dendrimer platform to achieve specific tumor targeting and siRNA-based gene silencing in combined and precision cancer therapy.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Pharmacy, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen, 518001, China
| | - Zhi Li
- Department of Pharmacy, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen, 518001, China
| | - Weiling Cao
- Department of Pharmacy, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen, 518001, China
| | - Jingjie Tang
- Aix-Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, "Equipe Labellisée Ligue Contre le Cancer", Marseille, 13288, France
| | - Yi Xia
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Ling Peng
- Aix-Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, "Equipe Labellisée Ligue Contre le Cancer", Marseille, 13288, France
| | - Jing Ma
- Department of Pharmacy, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| |
Collapse
|
12
|
Berggreen AH, Petersen JL, Lin L, Benabdellah K, Luo Y. CRISPR delivery with extracellular vesicles: Promises and challenges. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e111. [PMID: 38938376 PMCID: PMC11080907 DOI: 10.1002/jex2.111] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 06/29/2024]
Abstract
The CRISPR gene editing tool holds great potential for curing genetic disorders. However, the safe, efficient, and specific delivery of the CRISPR/Cas9 components into cells and tissues remains a challenge. While many currently available delivery methods achieve high levels of gene editing effects in vivo, they often result in genotoxicity and immunogenicity. Extracellular vesicles (EVs), which are cell-derived lipid nanoparticles, are capable of transferring protein and nucleic acid cargoes between cells, making them a promising endogenous alternative to synthetic delivery methods. This review provides a comprehensive analysis of the currently available strategies for EV-mediated delivery of CRISPR/Cas9. These strategies include cell-based, passive loading obtained by overexpression of CRISPR/Cas9, active loading involving protein or RNA dimerization, and loading into already purified EVs. All these approaches suggest that EV-based CRISPR/Cas9 delivery is useful for achieving both in vitro and in vivo gene editing. Despite that, substantial variations in cellular uptake and gene editing efficiencies indicate that further improvement and standardization are required for the therapeutic use of EVs as a CRISPR/Cas9 delivery vehicle. These improvements include, but is not limited to, the high-yield purification of EVs, increased loading and release efficiencies, as well as improved tissue- or cell-specific targeting specificities.
Collapse
Affiliation(s)
| | | | - Lin Lin
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Steno Diabetes Center AarhusAarhus University HospitalAarhusDenmark
| | - Karim Benabdellah
- Pfizer‐Universidad de Granada‐Junta de Andalucía Centre for Genomics and Oncological Research (GENYO)GranadaSpain
| | - Yonglun Luo
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Steno Diabetes Center AarhusAarhus University HospitalAarhusDenmark
- Lars Bolund Institute of Regenerative Medicine, Qingdao Europe Advanced Institute for Life SciencesBGI‐ResearchQingdaoChina
- Lars Bolund Institute of Regenerative Medicine, HIM‐BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesZhejiangHangzhouChina
| |
Collapse
|
13
|
Li Z, Lei Z, Cai Y, Cheng DB, Sun T. MicroRNA therapeutics and nucleic acid nano-delivery systems in bacterial infection: a review. J Mater Chem B 2023; 11:7804-7833. [PMID: 37539650 DOI: 10.1039/d3tb00694h] [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: 08/05/2023]
Abstract
Bacteria that have worked with humans for thousands of years pose a major threat to human health even today, as drug resistance has become a prominent problem. Compared to conventional drug therapy, nucleic acid-based therapies are a promising and potential therapeutic strategy for diseases in which nucleic acids are delivered through a nucleic acid delivery system to regulate gene expression in specific cells, offering the possibility of curing intractable diseases that are difficult to treat at this stage. Among the many nucleic acid therapeutic ideas, microRNA, a class of small nucleic acids with special properties, has made great strides in biology and medicine in just over two decades, showing promise in preclinical drug development. In this review, we introduce recent advances in nucleic acid delivery systems and their clinical applications, highlighting the potential of nucleic acid therapies, especially miRNAs extracted from traditional herbs, in combination with the existing set of nucleic acid therapeutic systems, to potentially open up a new line of thought in the treatment of cancer, viruses, and especially bacterial infectious diseases.
Collapse
Affiliation(s)
- Ze Li
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Yilun Cai
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| |
Collapse
|
14
|
Chen J, Zhu D, Lian B, Shi K, Chen P, Li Y, Lin W, Ding L, Long Q, Wang Y, Laurini E, Lan W, Li Y, Tintaru A, Ju C, Zhang C, Pricl S, Iovanna J, Liu X, Peng L. Cargo-selective and adaptive delivery of nucleic acid therapeutics by bola-amphiphilic dendrimers. Proc Natl Acad Sci U S A 2023; 120:e2220787120. [PMID: 37186846 PMCID: PMC10214173 DOI: 10.1073/pnas.2220787120] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/16/2023] [Indexed: 05/17/2023] Open
Abstract
Nucleic acid therapeutics are becoming an important drug modality, offering the unique opportunity to address "undruggable" targets, respond rapidly to evolving pathogens, and treat diseases at the gene level for precision medicine. However, nucleic acid therapeutics have poor bioavailability and are chemolabile and enzymolabile, imposing the need for delivery vectors. Dendrimers, by virtue of their well-defined structure and cooperative multivalence, represent precision delivery systems. We synthesized and studied bola-amphiphilic dendrimers for cargo-selective and on-demand delivery of DNA and small interfering RNA (siRNA), both important nucleic acid therapeutics. Remarkably, superior performances were achieved for siRNA delivery with the second-generation dendrimer, yet for DNA delivery with the third generation. We systematically studied these dendrimers with regard to cargo binding, cellular uptake, endosomal release, and in vivo delivery. Differences in size both of the dendrimers and their nucleic acid cargos impacted the cooperative multivalent interactions for cargo binding and release, leading to cargo-adaptive and selective delivery. Moreover, both dendrimers harnessed the advantages of lipid and polymer vectors, while offering nanotechnology-based tumor targeting and redox-responsive cargo release. Notably, they allowed tumor- and cancer cell-specific delivery of siRNA and DNA therapeutics for effective treatment in different cancer models, including aggressive and metastatic malignancies, outperforming the currently available vectors. This study provides avenues to engineer tailor-made vectors for nucleic acid delivery and precision medicine.
Collapse
Affiliation(s)
- Jiaxuan Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
- Aix Marseille University, CNRS, Center Interdisciplinaire de Nanoscience de Marseille, UMR 7325, «Equipe Labellisée Ligue Contre le Cancer», Marseille13288, France
| | - Dandan Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
| | - Baoping Lian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
| | - Kangjie Shi
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
| | - Peng Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
- Aix Marseille University, CNRS, Center Interdisciplinaire de Nanoscience de Marseille, UMR 7325, «Equipe Labellisée Ligue Contre le Cancer», Marseille13288, France
| | - Ying Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
| | - Wenyi Lin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
| | - Ling Ding
- Aix Marseille University, CNRS, Center Interdisciplinaire de Nanoscience de Marseille, UMR 7325, «Equipe Labellisée Ligue Contre le Cancer», Marseille13288, France
| | - Qiulin Long
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
| | - Yang Wang
- Aix Marseille University, CNRS, Center Interdisciplinaire de Nanoscience de Marseille, UMR 7325, «Equipe Labellisée Ligue Contre le Cancer», Marseille13288, France
- Hubei Gedian Humanwell Pharmaceutical Co. Ltd., E-zhou436070, P. R. China
| | - Erik Laurini
- Department of Engineering and Architecture, Molecular Biology and Nanotechnology Laboratory, University of Trieste, Trieste34127, Italy
| | - Wenjun Lan
- Aix Marseille University, CNRS, Center Interdisciplinaire de Nanoscience de Marseille, UMR 7325, «Equipe Labellisée Ligue Contre le Cancer», Marseille13288, France
- Aix Marseille University, INSERM, Centre de Recherche en Cancérologie de Marseille, Institute Pauli-Calmettes, Marseille13273, France
| | - Yun Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
| | - Aura Tintaru
- Aix Marseille University, CNRS, Center Interdisciplinaire de Nanoscience de Marseille, UMR 7325, «Equipe Labellisée Ligue Contre le Cancer», Marseille13288, France
| | - Caoyun Ju
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
| | - Sabrina Pricl
- Department of Engineering and Architecture, Molecular Biology and Nanotechnology Laboratory, University of Trieste, Trieste34127, Italy
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz90-136, Poland
| | - Juan Iovanna
- Aix Marseille University, INSERM, Centre de Recherche en Cancérologie de Marseille, Institute Pauli-Calmettes, Marseille13273, France
| | - Xiaoxuan Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing211198, P. R. China
| | - Ling Peng
- Aix Marseille University, CNRS, Center Interdisciplinaire de Nanoscience de Marseille, UMR 7325, «Equipe Labellisée Ligue Contre le Cancer», Marseille13288, France
| |
Collapse
|
15
|
Li D, Liu Q, Yang M, Xu H, Zhu M, Zhang Y, Xu J, Tian C, Yao J, Wang L, Liang Y. Nanomaterials for mRNA-based therapeutics: Challenges and opportunities. Bioeng Transl Med 2023; 8:e10492. [PMID: 37206219 PMCID: PMC10189457 DOI: 10.1002/btm2.10492] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/31/2023] Open
Abstract
Messenger RNA (mRNA) holds great potential in developing immunotherapy, protein replacement, and genome editing. In general, mRNA does not have the risk of being incorporated into the host genome and does not need to enter the nucleus for transfection, and it can be expressed even in nondividing cells. Therefore, mRNA-based therapeutics provide a promising strategy for clinical treatment. However, the efficient and safe delivery of mRNA remains a crucial constraint for the clinical application of mRNA therapeutics. Although the stability and tolerability of mRNA can be enhanced by directly retouching the mRNA structure, there is still an urgent need to improve the delivery of mRNA. Recently, significant progress has been made in nanobiotechnology, providing tools for developing mRNA nanocarriers. Nano-drug delivery system is directly used for loading, protecting, and releasing mRNA in the biological microenvironment and can be used to stimulate the translation of mRNA to develop effective intervention strategies. In the present review, we summarized the concept of emerging nanomaterials for mRNA delivery and the latest progress in enhancing the function of mRNA, primarily focusing on the role of exosomes in mRNA delivery. Moreover, we outlined its clinical applications so far. Finally, the key obstacles of mRNA nanocarriers are emphasized, and promising strategies to overcome these obstacles are proposed. Collectively, nano-design materials exert functions for specific mRNA applications, provide new perception for next-generation nanomaterials, and thus revolution of mRNA technology.
Collapse
Affiliation(s)
- De‐feng Li
- Department of GastroenterologyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Qi‐song Liu
- National Clinical Research Center for Infectious DiseasesShenzhen Third People's Hospital, Southern University of Science and TechnologyShenzhenChina
| | - Mei‐feng Yang
- Department of HematologyYantian District People's HospitalShenzhenGuangdongChina
| | - Hao‐ming Xu
- Department of Gastroenterology and HepatologyGuangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Min‐zheng Zhu
- Department of Gastroenterology and Hepatologythe Second Affiliated Hospital, School of Medicine, South China University of TechnologyGuangzhouGuangdongChina
| | - Yuan Zhang
- Department of Medical AdministrationHuizhou Institute of Occupational Diseases Control and PreventionHuizhouGuangdongChina
| | - Jing Xu
- Department of Gastroenterology and HepatologyGuangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Cheng‐mei Tian
- Department of EmergencyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Jun Yao
- Department of GastroenterologyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Li‐sheng Wang
- Department of GastroenterologyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Yu‐jie Liang
- Department of Child and Adolescent PsychiatryShenzhen Kangning Hospital, Shenzhen Mental Health CenterShenzhenChina
- Affiliated Hospital of Jining Medical University, Jining Medical UniversityJiningShandongChina
| |
Collapse
|
16
|
Galanakou C, Dhumal D, Peng L. Amphiphilic dendrimers against antibiotic resistance: light at the end of the tunnel? Biomater Sci 2023; 11:3379-3393. [PMID: 36866708 DOI: 10.1039/d2bm01878k] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
With the alarming and prevailing antimicrobial resistance (AMR) comes an urgent need for novel antimicrobial agents that are not only effective and robust but also do not induce resistance development. Amphiphilic dendrimers are emerging as a promising new paradigm to combat bacterial AMR. They can mimic antimicrobial peptides to produce potent antibacterial activity yet with a low likelihood of generating resistance. In addition, they are stable against enzymatic degradation thanks to their unique dendritic architecture. Importantly, these amphiphilic dendrimers are composed of distinct hydrophobic and hydrophilic entities bearing dendritic structures, which can be precisely designed and synthesized to optimize the hydrophobic-hydrophilic balance yielding potent antibacterial activity while minimizing adverse effects and drug resistance. In this short review, we present the challenges and current state of research in developing amphiphilic dendrimers as new antibiotic substitutes. We start with a brief overview of the advantages and opportunities associated with using amphiphilic dendrimers to combat bacterial AMR. We then outline the specific considerations and the mechanisms underlying the antibacterial activity of amphiphilic dendrimers. We focus on the importance of the amphiphilic nature of a dendrimer that balances hydrophobicity and hydrophilicity via gauging the hydrophobic entity and the dendrimer generation, branching unit, terminal group and charge to allow high antibacterial potency and selectivity while minimizing toxicity. Finally, we present the future challenges and perspectives for amphiphilic dendrimers as antibacterial candidates for combating AMR.
Collapse
Affiliation(s)
- Christina Galanakou
- Centre Interdisciplinaire de Nanoscience de Marseille, CINaM, UMR 7325, Aix Marseille University, CNRS, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France.
| | - Dinesh Dhumal
- Centre Interdisciplinaire de Nanoscience de Marseille, CINaM, UMR 7325, Aix Marseille University, CNRS, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France.
| | - Ling Peng
- Centre Interdisciplinaire de Nanoscience de Marseille, CINaM, UMR 7325, Aix Marseille University, CNRS, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France.
| |
Collapse
|
17
|
Jiang Y, Lyu Z, Ralahy B, Liu J, Roussel T, Ding L, Tang J, Kosta A, Giorgio S, Tomasini R, Liang XJ, Dusetti N, Iovanna J, Peng L. Dendrimer nanosystems for adaptive tumor-assisted drug delivery via extracellular vesicle hijacking. Proc Natl Acad Sci U S A 2023; 120:e2215308120. [PMID: 36745793 PMCID: PMC9963653 DOI: 10.1073/pnas.2215308120] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/04/2023] [Indexed: 02/08/2023] Open
Abstract
Drug delivery systems (DDSs) that can overcome tumor heterogeneity and achieve deep tumor penetration are challenging to develop yet in high demand for cancer treatment. We report here a DDS based on self-assembling dendrimer nanomicelles for effective and deep tumor penetration via in situ tumor-secreted extracellular vesicles (EVs), an endogenous transport system that evolves with tumor microenvironment. Upon arrival at a tumor, these dendrimer nanomicelles had their payload repackaged by the cells into EVs, which were further transported and internalized by other cells for delivery "in relay." Using pancreatic and colorectal cancer-derived 2D, 3D, and xenograft models, we demonstrated that the in situ-generated EVs mediated intercellular delivery, propagating cargo from cell to cell and deep within the tumor. Our study provides a new perspective on exploiting the intrinsic features of tumors alongside dendrimer supramolecular chemistry to develop smart and effective DDSs to overcome tumor heterogeneity and their evolutive nature thereby improving cancer therapy.
Collapse
Affiliation(s)
- Yifan Jiang
- Aix Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288France
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS, UMR 7258, Institut Paoli-Calmettes, Aix Marseille Université, 13273Marseille, France
| | - Zhenbin Lyu
- Aix Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288France
| | - Brigino Ralahy
- Aix Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288France
| | - Juan Liu
- Aix Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288France
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, 100190Beijing, China
- University of Chinese Academy of Sciences, 100049Beijing, China
| | - Tom Roussel
- Aix Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288France
| | - Ling Ding
- Aix Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288France
| | - Jingjie Tang
- Aix Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288France
| | - Artemis Kosta
- Aix Marseille Université, CNRS, Mediterranean Institute of Microbiology,FR3479, 13009Marseille, France
| | - Suzanne Giorgio
- Aix Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288France
| | - Richard Tomasini
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS, UMR 7258, Institut Paoli-Calmettes, Aix Marseille Université, 13273Marseille, France
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, 100190Beijing, China
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS, UMR 7258, Institut Paoli-Calmettes, Aix Marseille Université, 13273Marseille, France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS, UMR 7258, Institut Paoli-Calmettes, Aix Marseille Université, 13273Marseille, France
| | - Ling Peng
- Aix Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, 13288France
| |
Collapse
|
18
|
Yu Z, Tsapis N, Fay F, Chen L, Karpus A, Shi X, Cailleau C, García Pérez S, Huang N, Vergnaud J, Mignani S, Majoral JP, Fattal E. Amphiphilic Phosphorus Dendrons Associated with Anti-inflammatory siRNA Reduce Symptoms in Murine Collagen-Induced Arthritis. Biomacromolecules 2023; 24:667-677. [PMID: 36599673 DOI: 10.1021/acs.biomac.2c01117] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Small interfering RNA (siRNA) holds promise for treating rheumatoid arthritis by inhibiting major cytokines such as tumor necrosis factor-α (TNF-α). We developed original cationic amphiphilic phosphorus dendrons to produce dendriplexes associated with TNF-α siRNA. The dendrons were made of 10 pyrrolidinium end groups and a C17 aliphatic chain. The dendriplexes demonstrated the ability to protect siRNA from nuclease degradation and to promote macrophage uptake. Moreover, they led to potent inhibition of TNF-α expression in the lipopolysaccharide-activated mouse macrophage cell line RAW264.7 in vitro model. A significant anti-inflammatory effect in the murine collagen-induced arthritis model was observed through arthritis scoring and histological observations. These results open up essential perspectives in using this original amphiphilic dendron to reduce the disease burden and improve outcomes in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Zhibo Yu
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - Nicolas Tsapis
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - François Fay
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - Liang Chen
- Laboratoire de Chimie de Coordination, CNRS, Université de Toulouse, Toulouse Cedex 431077, France.,Université Toulouse, 118 Route de Narbonne, Toulouse Cedex431077, France
| | - Andrii Karpus
- Laboratoire de Chimie de Coordination, CNRS, Université de Toulouse, Toulouse Cedex 431077, France.,Université Toulouse, 118 Route de Narbonne, Toulouse Cedex431077, France
| | - Xiangyang Shi
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai201620, PR China
| | - Catherine Cailleau
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - Samuel García Pérez
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), Sergas-Uvigo36213, Spain
| | - Nicolas Huang
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - Juliette Vergnaud
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| | - Serge Mignani
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologique, University Paris Descartes, Pres Sorbonne Paris Cité, CNRS UMR 860, 45 Rue des Saints Peres, Paris75006, France.,CQM─Centro de Química da Madeira, Universidade da Madeira, Funchal9020-105, Portugal
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination, CNRS, Université de Toulouse, Toulouse Cedex 431077, France
| | - Elias Fattal
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Orsay91400, France
| |
Collapse
|
19
|
Căta A, Ienașcu IMC, Ştefănuț MN, Roșu D, Pop OR. Properties and Bioapplications of Amphiphilic Janus Dendrimers: A Review. Pharmaceutics 2023; 15:589. [PMID: 36839911 PMCID: PMC9958631 DOI: 10.3390/pharmaceutics15020589] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Amphiphilic Janus dendrimers are arrangements containing both hydrophilic and hydrophobic units, capable of forming ordered aggregates by intermolecular noncovalent interactions between the dendrimer units. Compared to conventional dendrimers, these molecular self-assemblies possess particular and effective attributes i.e., the presence of different terminal groups, essential to design new elaborated materials. The present review will focus on the pharmaceutical and biomedical application of amphiphilic Janus dendrimers. Important information for the development of novel optimized pharmaceutical formulations, such as structural classification, synthetic pathways, properties and applications, will offer the complete characterization of this type of Janus dendrimers. This work will constitute an up-to-date background for dendrimer specialists involved in designing amphiphilic Janus dendrimer-based nanomaterials for future innovations in this promising field.
Collapse
Affiliation(s)
- Adina Căta
- National Institute of Research and Development for Electrochemistry and Condensed Matter, 144 Dr. A. P. Podeanu, 300569 Timişoara, Romania
| | - Ioana Maria Carmen Ienașcu
- National Institute of Research and Development for Electrochemistry and Condensed Matter, 144 Dr. A. P. Podeanu, 300569 Timişoara, Romania
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, “Vasile Goldiș” Western University of Arad, 86 Liviu Rebreanu, 310045 Arad, Romania
| | - Mariana Nela Ştefănuț
- National Institute of Research and Development for Electrochemistry and Condensed Matter, 144 Dr. A. P. Podeanu, 300569 Timişoara, Romania
| | - Dan Roșu
- National Institute of Research and Development for Electrochemistry and Condensed Matter, 144 Dr. A. P. Podeanu, 300569 Timişoara, Romania
| | - Oana-Raluca Pop
- Faculty of Pharmacy, University of Medicine and Pharmacy “Victor Babeș” Timișoara, 2 Eftimie Murgu Square, 300041 Timișoara, Romania
| |
Collapse
|
20
|
Brito ME, Mikhtaniuk SE, Neelov IM, Borisov OV, Holm C. Implicit-Solvent Coarse-Grained Simulations of Linear-Dendritic Block Copolymer Micelles. Int J Mol Sci 2023; 24:2763. [PMID: 36769091 PMCID: PMC9917066 DOI: 10.3390/ijms24032763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
The design of nanoassemblies can be conveniently achieved by tuning the strength of the hydrophobic interactions of block copolymers in selective solvents. These block copolymer micelles form supramolecular aggregates, which have attracted great attention in the area of drug delivery and imaging in biomedicine due to their easy-to-tune properties and straightforward large-scale production. In the present work, we have investigated the micellization process of linear-dendritic block copolymers in order to elucidate the effect of branching on the micellar properties. We focus on block copolymers formed by linear hydrophobic blocks attached to either dendritic neutral or charged hydrophilic blocks. We have implemented a simple protocol for determining the equilibrium micellar size, which permits the study of linear-dendritic block copolymers in a wide range of block morphologies in an efficient and parallelizable manner. We have explored the impact of different topological and charge properties of the hydrophilic blocks on the equilibrium micellar properties and compared them to predictions from self-consistent field theory and scaling theory. We have found that, at higher degrees of branching in the corona and for short polymer chains, excluded volume interactions strongly influence the micellar aggregation as well as their effective charge.
Collapse
Affiliation(s)
- Mariano E. Brito
- Institute for Computational Physics, University of Stuttgart, D-70569 Stuttgart, Germany
| | - Sofia E. Mikhtaniuk
- School of Computer Technologies and Control, St. Petersburg National Research University of Information Technologies, Mechanics and Optics, 197101 St. Petersburg, Russia
| | - Igor M. Neelov
- School of Computer Technologies and Control, St. Petersburg National Research University of Information Technologies, Mechanics and Optics, 197101 St. Petersburg, Russia
| | - Oleg V. Borisov
- School of Computer Technologies and Control, St. Petersburg National Research University of Information Technologies, Mechanics and Optics, 197101 St. Petersburg, Russia
- Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux, UMR 5254 CNRS UPPA, 64053 Pau, France
| | - Christian Holm
- Institute for Computational Physics, University of Stuttgart, D-70569 Stuttgart, Germany
| |
Collapse
|
21
|
Effect of amphiphilic phosphorous dendrons on the conformation, secondary structure, and zeta potential of albumin and thrombin. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04512-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Xiong Y, Ke R, Zhang Q, Lan W, Yuan W, Chan KNI, Roussel T, Jiang Y, Wu J, Liu S, Wong AST, Shim JS, Zhang X, Xie R, Dusetti N, Iovanna J, Habib N, Peng L, Lee LTO. Small Activating RNA Modulation of the G Protein-Coupled Receptor for Cancer Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200562. [PMID: 35712764 PMCID: PMC9475523 DOI: 10.1002/advs.202200562] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/23/2022] [Indexed: 06/15/2023]
Abstract
G protein-coupled receptors (GPCRs) are the most common and important drug targets. However, >70% of GPCRs are undruggable or difficult to target using conventional chemical agonists/antagonists. Small nucleic acid molecules, which can sequence-specifically modulate any gene, offer a unique opportunity to effectively expand drug targets, especially those that are undruggable or difficult to address, such as GPCRs. Here, the authors report for the first time that small activating RNAs (saRNAs) effectively modulate a GPCR for cancer treatment. Specifically, saRNAs promoting the expression of Mas receptor (MAS1), a GPCR that counteracts the classical angiotensin II pathway in cancer cell proliferation and migration, are identified. These saRNAs, delivered by an amphiphilic dendrimer vector, enhance MAS1 expression, counteracting the angiotensin II/angiotensin II Receptor Type 1 axis, and leading to significant suppression of tumorigenesis and the inhibition of tumor progression of multiple cancers in tumor-xenografted mouse models and patient-derived tumor models. This study provides not only a new strategy for cancer therapy by targeting the renin-angiotensin system, but also a new avenue to modulate GPCR signaling by RNA activation.
Collapse
Affiliation(s)
- Yunfang Xiong
- Cancer CentreFaculty of Health SciencesUniversity of MacauTaipaMacau999078China
| | - Ran Ke
- Cancer CentreFaculty of Health SciencesUniversity of MacauTaipaMacau999078China
| | - Qingyu Zhang
- Department of Obstetrics and GynaecologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong524001China
| | - Wenjun Lan
- Aix Marseille UniversitéCNRSCentre Interdisciplinaire de Nanoscience de Marseille (UMR 7325)Equipe Labellisée Ligue Contre le CancerMarseille13288France
- Centre de Recherche en Cancérologie de Marseille (CRCM)INSERM U1068CNRSAix‐Marseille Université and Institut Paoli‐CalmettesMarseille13288France
| | - Wanjun Yuan
- Cancer CentreFaculty of Health SciencesUniversity of MacauTaipaMacau999078China
| | - Karol Nga Ieng Chan
- Cancer CentreFaculty of Health SciencesUniversity of MacauTaipaMacau999078China
| | - Tom Roussel
- Aix Marseille UniversitéCNRSCentre Interdisciplinaire de Nanoscience de Marseille (UMR 7325)Equipe Labellisée Ligue Contre le CancerMarseille13288France
| | - Yifan Jiang
- Aix Marseille UniversitéCNRSCentre Interdisciplinaire de Nanoscience de Marseille (UMR 7325)Equipe Labellisée Ligue Contre le CancerMarseille13288France
| | - Jing Wu
- Aix Marseille UniversitéCNRSCentre Interdisciplinaire de Nanoscience de Marseille (UMR 7325)Equipe Labellisée Ligue Contre le CancerMarseille13288France
| | - Shuai Liu
- Cancer CentreFaculty of Health SciencesUniversity of MacauTaipaMacau999078China
| | - Alice Sze Tsai Wong
- School of Biological SciencesThe University of Hong KongPokfulam RoadHong KongChina
| | - Joong Sup Shim
- Cancer CentreFaculty of Health SciencesUniversity of MacauTaipaMacau999078China
- MOE Frontiers Science Center for Precision OncologyUniversity of MacauTaipaMacau999078China
| | - Xuanjun Zhang
- Cancer CentreFaculty of Health SciencesUniversity of MacauTaipaMacau999078China
- MOE Frontiers Science Center for Precision OncologyUniversity of MacauTaipaMacau999078China
| | - Ruiyu Xie
- Cancer CentreFaculty of Health SciencesUniversity of MacauTaipaMacau999078China
- MOE Frontiers Science Center for Precision OncologyUniversity of MacauTaipaMacau999078China
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM)INSERM U1068CNRSAix‐Marseille Université and Institut Paoli‐CalmettesMarseille13288France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM)INSERM U1068CNRSAix‐Marseille Université and Institut Paoli‐CalmettesMarseille13288France
| | - Nagy Habib
- Department of Surgery and CancerImperial College LondonLondonW12 0NNUK
- MiNA Therapeutics, Translation & Innovation Hub80 Wood LaneLondonW12 0BZUK
| | - Ling Peng
- Aix Marseille UniversitéCNRSCentre Interdisciplinaire de Nanoscience de Marseille (UMR 7325)Equipe Labellisée Ligue Contre le CancerMarseille13288France
| | - Leo Tsz On Lee
- Cancer CentreFaculty of Health SciencesUniversity of MacauTaipaMacau999078China
- MOE Frontiers Science Center for Precision OncologyUniversity of MacauTaipaMacau999078China
- Centre of Reproduction, Development, and AgingFaculty of Health SciencesUniversity of MacauTaipaMacau999078China
| |
Collapse
|
23
|
Bera S, Barman R, Ghosh S. Hyperbranched vs. linear poly(disulfide) for intracellular drug delivery. Polym Chem 2022. [DOI: 10.1039/d2py00896c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This communication reports comparative studies between amphiphilic hyperbranched and linear poly(disulfide) with regard to their aggregation and glutathione-responsive intracellular drug delivery.
Collapse
Affiliation(s)
- Sukanya Bera
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata, India-700032
| | - Ranajit Barman
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata, India-700032
| | - Suhrit Ghosh
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata, India-700032
| |
Collapse
|