1
|
Nasr Azadani M, Abed A, Mirzaei SA, Mahjoubin-Tehran M, Hamblin M, Rahimian N, Mirzaei H. Nanoparticles in Cancer Theranostics: Focus on Gliomas. BIONANOSCIENCE 2025; 15:129. [DOI: 10.1007/s12668-024-01752-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 01/05/2025]
|
2
|
Khramtsov YV, Bunin ES, Ulasov AV, Lupanova TN, Georgiev GP, Sobolev AS. GALA3-Containing Modular Nanotransporters Are Capable of Delivering Keap1 Monobody to Target Cells and Inhibiting the Formation of Reactive Oxygen Species in the Cells. DOKL BIOCHEM BIOPHYS 2025:10.1134/S1607672924601252. [PMID: 39899249 DOI: 10.1134/s1607672924601252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 02/04/2025]
Abstract
In the previously created modular nanotransporter (MNT) capable of delivering a monobody to Keap1 into the cytosol, the endosomolytic module, translocation domain of diphtheria toxin (DTox), was replaced by the endosomolytic peptide GALA3. It was found that this substitution more than doubles the lifetime of MNT in the blood. Using confocal microscopy, it was shown that MNT with GALA3 was internalized into AML12 cells mainly due to binding to the epidermal growth factor receptor, and is also able to exit from endosomes into the cytosol. Using cellular thermal shift assay, it was shown that MNT with GALA3 and MNT with DTox are equally effective in disrupting the formation of the Nrf2 complex with Keap1, which led to similar protection of AML12 cells from the action of hydrogen peroxide. The obtained results allow not only optimizing the systemic use of MNT, but can also serve as a basis for creating agents aimed at treating diseases associated with oxidative stress.
Collapse
Affiliation(s)
| | - E S Bunin
- Institute of Gene Biology, RAS, Moscow, Russia
- Moscow State University, Moscow, Russia
| | - A V Ulasov
- Institute of Gene Biology, RAS, Moscow, Russia
| | | | | | - A S Sobolev
- Institute of Gene Biology, RAS, Moscow, Russia.
- Moscow State University, Moscow, Russia.
| |
Collapse
|
3
|
Wang H, Hsu JC, Song W, Lan X, Cai W, Ni D. Nanorepair medicine for treatment of organ injury. Natl Sci Rev 2024; 11:nwae280. [PMID: 39257435 PMCID: PMC11384914 DOI: 10.1093/nsr/nwae280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/24/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024] Open
Abstract
Organ injuries, such as acute kidney injury, ischemic stroke, and spinal cord injury, often result in complications that can be life-threatening or even fatal. Recently, many nanomaterials have emerged as promising agents for repairing various organ injuries. In this review, we present the important developments in the field of nanomaterial-based repair medicine, herein referred to as 'nanorepair medicine'. We first introduce the disease characteristics associated with different types of organ injuries and highlight key examples of relevant nanorepair medicine. We then provide a summary of existing strategies in nanorepair medicine, including organ-targeting methodologies and potential countermeasures against exogenous and endogenous pathologic risk factors. Finally, we offer our perspectives on current challenges and future expectations for the advancement of nanomedicine designed for organ injury repair.
Collapse
Affiliation(s)
- Han Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Wenyu Song
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430073, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan 430073, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430073, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan 430073, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dalong Ni
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
4
|
Huo CM, Zuo YC, Chen Y, Chen L, Zhu JY, Xue W. Natural lignin nanoparticles target tumor by saturating the phagocytic capacity of Kupffer cells in the liver. Int J Biol Macromol 2024; 274:133186. [PMID: 38885858 DOI: 10.1016/j.ijbiomac.2024.133186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
Ligand-receptor recognition serves as the fundamental driving force for active targeting, yet it is still constrained by off-target effects. Herein, we demonstrate that circumventing or blocking the mononuclear phagocyte system (MPS) are both viable strategies to address off-target effects. Naturally derived lignin nanoparticles (LNPs) show great potential to block MPS due to its good stability, low toxicity, and degradability. We further demonstrate the impact of LNPs dosage on in vivo tumor targeting and antitumor efficacy. Our results show that a high dose of LNPs (300 mg/kg) leads to significant accumulation at the tumor site for a duration of 14 days after intravenous administration. In contrast, the low-dose counterparts (e.g., 50, 150 mg/kg) result in almost all LNPs accumulating in the liver. This discovery indicates that the liver is the primary site of LNP capture, leaving only the surplus LNPs the chance to reach the tumor. In addition, although cell membrane-engineered LNPs can rapidly penetrate tumors, they are still prone to capture by the liver during subsequent circulation in the bloodstream. Excitingly, comparable therapeutic efficacy is obtained for the above two strategies. Our findings may offer valuable insights into the targeted delivery of drugs for disease treatment.
Collapse
Affiliation(s)
- Cong-Min Huo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yu-Cheng Zuo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yu Chen
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Liheng Chen
- Guangdong Provincial Key Laboratory of Plant Resources Biorefinery, School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China.
| | - Jing-Yi Zhu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
5
|
Kolesnikova OA, Komedchikova EN, Zvereva SD, Obozina AS, Dorozh OV, Afanasev I, Nikitin PI, Mochalova EN, Nikitin MP, Shipunova VO. Albumin incorporation into recognising layer of HER2-specific magnetic nanoparticles as a tool for optimal targeting of the acidic tumor microenvironment. Heliyon 2024; 10:e34211. [PMID: 39100472 PMCID: PMC11296017 DOI: 10.1016/j.heliyon.2024.e34211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 07/05/2024] [Indexed: 08/06/2024] Open
Abstract
Cancer is unquestionably a global healthcare challenge, spurring the exporation of novel treatment approaches. In recent years, nanomaterials have garnered significant interest with the greatest hopes for targeted nanoformulations due to their cell-specific delivery, improved therapeutic efficacy, and reduced systemic toxicity for the organism. The problem of successful clinical translation of nanoparticles may be related to the fact that most in vitro tests are performed at pH values of normal cells and tissues, ranging from 7.2 to 7.4. The extracellular pH values of tumors are characterized by a shift to a more acidic region in the range of 5.6-7.0 and represent a crucial target for enhancing nanoparticle delivery to cancer cells. Here we show the method of non-active protein incorporation into the surface of HER2-targeted nanoparticles to achieve optimal cellular uptake within the pH range of the tumor microenvironment. The method efficacy was confirmed in vitro and in vivo showing the maximum binding of nanoparticles to cells at a pH value 6.4. Namely, fluorescent magnetic nanoparticles, modified with HER2-recognising affibody ZHER2:342, with proven specificity in terms of HER2 recognition (with 62-fold higher cellular uptake compared to control nanoparticles) were designed for targeting cancer cells at slightly acidic pH values. The stabilizing protein, namely, bovine serum albumin, one of the major blood components with widespread availability and biocompatibility, was used for the decoration of the nanoparticle surface to alter the pH response of the targeting magnetic conjugates. The optimally designed nanoparticles showed a bell-shaped dependency of interaction with cancer cells in the pH range of 5.6-8.0 with maximum cellular uptake at pH value 6.4 close to that of the tumor microenvironment. In vivo experiments revealed that after i.v. administration, BSA-decorated nanoparticles exhibited 2 times higher accumulation in tumors compared to magnetic nanoparticles modified with affibody only. Thus, we demonstrated a valid method for enhancing the specificity of targeted nanoparticle delivery to cancer cells without changing the functional components of nanoparticles.
Collapse
Affiliation(s)
- Olga A. Kolesnikova
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
| | - Elena N. Komedchikova
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701, Dolgoprudny, Russia
| | - Svetlana D. Zvereva
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701, Dolgoprudny, Russia
| | | | - Olha V. Dorozh
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
| | - Iurii Afanasev
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701, Dolgoprudny, Russia
| | - Petr I. Nikitin
- Prokhorov General Physics Institute, Russian Academy of Sciences, 38 Vavilov Street, 119991, Moscow, Russia
| | - Elizaveta N. Mochalova
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Department of Nanobiomedicine, Sirius University of Science and Technology, 1 Olympic Ave., 354340, Sochi, Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701, Dolgoprudny, Russia
| | - Maxim P. Nikitin
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Department of Nanobiomedicine, Sirius University of Science and Technology, 1 Olympic Ave., 354340, Sochi, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia
| | - Victoria O. Shipunova
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Department of Nanobiomedicine, Sirius University of Science and Technology, 1 Olympic Ave., 354340, Sochi, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia
| |
Collapse
|
6
|
Wang Y, Liu C, Ren Y, Song J, Fan K, Gao L, Ji X, Chen X, Zhao H. Nanomaterial-Based Strategies for Attenuating T-Cell-Mediated Immunodepression in Stroke Patients: Advancing Research Perspectives. Int J Nanomedicine 2024; 19:5793-5812. [PMID: 38882535 PMCID: PMC11180442 DOI: 10.2147/ijn.s456632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
This review article discusses the potential of nanomaterials in targeted therapy and immunomodulation for stroke-induced immunosuppression. Although nanomaterials have been extensively studied in various biomedical applications, their specific use in studying and addressing immunosuppression after stroke remains limited. Stroke-induced neuroinflammation is characterized by T-cell-mediated immunodepression, which leads to increased morbidity and mortality. Key observations related to immunodepression after stroke, including lymphopenia, T-cell dysfunction, regulatory T-cell imbalance, and cytokine dysregulation, are discussed. Nanomaterials, such as liposomes, micelles, polymeric nanoparticles, and dendrimers, offer advantages in the precise delivery of drugs to T cells, enabling enhanced targeting and controlled release of immunomodulatory agents. These nanomaterials have the potential to modulate T-cell function, promote neuroregeneration, and restore immune responses, providing new avenues for stroke treatment. However, challenges related to biocompatibility, stability, scalability, and clinical translation need to be addressed. Future research efforts should focus on comprehensive studies to validate the efficacy and safety of nanomaterial-based interventions targeting T cells in stroke-induced immunosuppression. Collaborative interdisciplinary approaches are necessary to advance the field and translate these innovative strategies into clinical practice, ultimately improving stroke outcomes and patient care.
Collapse
Grants
- This work was supported by the National Natural Science Foundation of China (Grant number 82001248), National University of Singapore (NUHSRO/2020/133/Startup/08, NUHSRO/2023/008/NUSMed/TCE/LOA, NUHSRO/2021/034/TRP/09/Nanomedicine, NUHSRO/2021/044/Kickstart/09/LOA, 23-0173-A0001), National Medical Research Council (MOH-001388-00, CG21APR1005, OFIRG23jul-0047), Singapore Ministry of Education (MOE-000387-00), and National Research Foundation (NRF-000352-00)
Collapse
Affiliation(s)
- Yan Wang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Cuiying Liu
- School of Nursing, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Yanhong Ren
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Heng Zhao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
7
|
Liao J, Gong L, Xu Q, Wang J, Yang Y, Zhang S, Dong J, Lin K, Liang Z, Sun Y, Mu Y, Chen Z, Lu Y, Zhang Q, Lin Z. Revolutionizing Neurocare: Biomimetic Nanodelivery Via Cell Membranes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402445. [PMID: 38583077 DOI: 10.1002/adma.202402445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Brain disorders represent a significant challenge in medical science due to the formidable blood-brain barrier (BBB), which severely limits the penetration of conventional therapeutics, hindering effective treatment strategies. This review delves into the innovative realm of biomimetic nanodelivery systems, including stem cell-derived nanoghosts, tumor cell membrane-coated nanoparticles, and erythrocyte membrane-based carriers, highlighting their potential to circumvent the BBB's restrictions. By mimicking native cell properties, these nanocarriers emerge as a promising solution for enhancing drug delivery to the brain, offering a strategic advantage in overcoming the barrier's selective permeability. The unique benefits of leveraging cell membranes from various sources is evaluated and advanced technologies for fabricating cell membrane-encapsulated nanoparticles capable of masquerading as endogenous cells are examined. This enables the targeted delivery of a broad spectrum of therapeutic agents, ranging from small molecule drugs to proteins, thereby providing an innovative approach to neurocare. Further, the review contrasts the capabilities and limitations of these biomimetic nanocarriers with traditional delivery methods, underlining their potential to enable targeted, sustained, and minimally invasive treatment modalities. This review is concluded with a perspective on the clinical translation of these biomimetic systems, underscoring their transformative impact on the therapeutic landscape for intractable brain diseases.
Collapse
Affiliation(s)
- Jun Liao
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Lidong Gong
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Qingqiang Xu
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Jingya Wang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yuanyuan Yang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Shiming Zhang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Junwei Dong
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Kerui Lin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Zichao Liang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yuhan Sun
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Yongxu Mu
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014040, China
| | - Zhengju Chen
- Pooling Medical Research Institutes of 100Biotech, Beijing, 100006, China
| | - Ying Lu
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Qiang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zhiqiang Lin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
8
|
Zelepukin IV, Shevchenko KG, Deyev SM. Rediscovery of mononuclear phagocyte system blockade for nanoparticle drug delivery. Nat Commun 2024; 15:4366. [PMID: 38777821 PMCID: PMC11111695 DOI: 10.1038/s41467-024-48838-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
Rapid uptake of nanoparticles by mononuclear phagocyte system (MPS) significantly hampers their therapeutic efficacy. Temporal MPS blockade is one of the few ways to overcome this barrier - the approach rediscovered many times under different names but never extensively used in clinic. Using meta-analysis of the published data we prove the efficacy of this technique for enhancing particle circulation in blood and their delivery to tumours, describe a century of its evolution and potential combined mechanism behind it. Finally, we discuss future directions of the research focusing on the features essential for successful clinical translation of the method.
Collapse
Affiliation(s)
- Ivan V Zelepukin
- Department of Medicinal Chemistry, Uppsala University, 751 23, Uppsala, Sweden.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia.
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia
| |
Collapse
|
9
|
Piotrowski-Daspit AS, Bracaglia LG, Eaton DA, Richfield O, Binns TC, Albert C, Gould J, Mortlock RD, Egan ME, Pober JS, Saltzman WM. Enhancing in vivo cell and tissue targeting by modulation of polymer nanoparticles and macrophage decoys. Nat Commun 2024; 15:4247. [PMID: 38762483 PMCID: PMC11102454 DOI: 10.1038/s41467-024-48442-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 05/01/2024] [Indexed: 05/20/2024] Open
Abstract
The in vivo efficacy of polymeric nanoparticles (NPs) is dependent on their pharmacokinetics, including time in circulation and tissue tropism. Here we explore the structure-function relationships guiding physiological fate of a library of poly(amine-co-ester) (PACE) NPs with different compositions and surface properties. We find that circulation half-life as well as tissue and cell-type tropism is dependent on polymer chemistry, vehicle characteristics, dosing, and strategic co-administration of distribution modifiers, suggesting that physiological fate can be optimized by adjusting these parameters. Our high-throughput quantitative microscopy-based platform to measure the concentration of nanomedicines in the blood combined with detailed biodistribution assessments and pharmacokinetic modeling provides valuable insight into the dynamic in vivo behavior of these polymer NPs. Our results suggest that PACE NPs-and perhaps other NPs-can be designed with tunable properties to achieve desired tissue tropism for the in vivo delivery of nucleic acid therapeutics. These findings can guide the rational design of more effective nucleic acid delivery vehicles for in vivo applications.
Collapse
Affiliation(s)
- Alexandra S Piotrowski-Daspit
- Department of Biomedical Engineering, Yale University, New Haven, CT, US.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, US.
- Department of Internal Medicine - Pulmonary and Critical Care Medicine Division, Michigan Medicine, University of Michigan, Ann Arbor, MI, US.
| | - Laura G Bracaglia
- Department of Biomedical Engineering, Yale University, New Haven, CT, US.
- Department of Chemical and Biological Engineering, Villanova University, Villanova, PA, US.
| | - David A Eaton
- Department of Biomedical Engineering, Yale University, New Haven, CT, US
| | - Owen Richfield
- Department of Biomedical Engineering, Yale University, New Haven, CT, US
| | - Thomas C Binns
- Department of Biomedical Engineering, Yale University, New Haven, CT, US
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, US
| | - Claire Albert
- Department of Biomedical Engineering, Yale University, New Haven, CT, US
| | - Jared Gould
- Department of Biomedical Engineering, Yale University, New Haven, CT, US
| | - Ryland D Mortlock
- Department of Biomedical Engineering, Yale University, New Haven, CT, US
| | - Marie E Egan
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, US
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, US
| | - Jordan S Pober
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, US
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, US
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, US.
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, US.
- Department of Dermatology, Yale School of Medicine, New Haven, CT, US.
- Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, US.
| |
Collapse
|
10
|
Tan G, Hou G, Qian J, Wang Y, Xu W, Luo W, Chen X, Suo A. Hyaluronan-decorated copper-doxorubicin-anlotinib nanoconjugate for targeted synergistic chemo/chemodynamic/antiangiogenic tritherapy against hepatocellular carcinoma. J Colloid Interface Sci 2024; 662:857-869. [PMID: 38382370 DOI: 10.1016/j.jcis.2024.02.085] [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: 01/24/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Copper-based nanomaterials show considerable potential in the chemodynamic therapy of cancers. However, their clinical application is restricted by low catalytic activity in tumor microenvironment and copper-induced tumor angiogenesis. Herein, a novel copper-doxorubicin-anlotinib (CDA) nanoconjugate was constructed by the combination of copper-hydrazide coordination, hydrazone linkage and Schiff base bond. The CDA nanoconjugate consists of a copper-3,3'-dithiobis(propionohydrazide)-doxorubicin core and an anlotinib-hyaluronan shell. Benefiting from hyaluronan camouflage and abundant disulfide bonds and Cu2+, the CDA nanoconjugate possessed excellent tumor-targeting and glutathione-depleting abilities and enhanced chemodynamic efficacy. Released doxorubicin significantly improved copper-mediated chemodynamic therapy by upregulating nicotinamide adenine dinucleotide phosphate oxidase 4 expression to increase intracellular H2O2 level. Furthermore, the nanoconjugate produced excessive •OH to induce lipid peroxidation and mitochondrial dysfunction, thus greatly elevating doxorubicin-mediated chemotherapy. Importantly, anlotinib effectively inhibited the angiogenic potential of copper ions. In a word, the CDA nanoconjugate is successfully constructed by combined coordination and pH-responsive linkages, and displays the great potential of copper-drug conjugate for targeted synergistic chemo/chemodynamic/antiangiogenic triple therapy against cancers.
Collapse
Affiliation(s)
- Gang Tan
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Guanghui Hou
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China; Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Junmin Qian
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Yaping Wang
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Weijun Xu
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Wenjuan Luo
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaobing Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Aili Suo
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
11
|
Gabashvili AN, Alexandrushkina NA, Mochalova EN, Goliusova DV, Sapozhnikova EN, Makarevich PI, Nikitin PI. Internalization of transferrin-tagged Myxococcus xanthus encapsulins into mesenchymal stem cells. Exp Biol Med (Maywood) 2024; 249:10055. [PMID: 38774281 PMCID: PMC11106444 DOI: 10.3389/ebm.2024.10055] [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: 11/20/2023] [Accepted: 04/23/2024] [Indexed: 05/24/2024] Open
Abstract
Currently, various functionalized nanocarrier systems are extensively studied for targeted delivery of drugs, peptides, and nucleic acids. Joining the approaches of genetic and chemical engineering may produce novel carriers for precise targeting different cellular proteins, which is important for both therapy and diagnosis of various pathologies. Here we present the novel nanocontainers based on vectorized genetically encoded Myxococcus xanthus (Mx) encapsulin, confining a fluorescent photoactivatable mCherry (PAmCherry) protein. The shells of such encapsulins were modified using chemical conjugation of human transferrin (Tf) prelabeled with a fluorescein-6 (FAM) maleimide acting as a vector. We demonstrate that the vectorized encapsulin specifically binds to transferrin receptors (TfRs) on the membranes of mesenchymal stromal/stem cells (MSCs) followed by internalization into cells. Two spectrally separated fluorescent signals from Tf-FAM and PAmCherry are clearly distinguishable and co-localized. It is shown that Tf-tagged Mx encapsulins are internalized by MSCs much more efficiently than by fibroblasts. It has been also found that unlabeled Tf effectively competes with the conjugated Mx-Tf-FAM formulations. That indicates the conjugate internalization into cells by Tf-TfR endocytosis pathway. The developed nanoplatform can be used as an alternative to conventional nanocarriers for targeted delivery of, e.g., genetic material to MSCs.
Collapse
Affiliation(s)
- Anna N. Gabashvili
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, Russia
| | - Natalya A. Alexandrushkina
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Elizaveta N. Mochalova
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, Russia
- Moscow Center for Advanced Studies, Moscow, Russia
- Nanobiomedicine Division, Sirius University of Science and Technology, Sirius, Russia
| | - Daria V. Goliusova
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
- Laboratory of Cell Biology, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of FMBA, Moscow, Russia
| | | | - Pavel I. Makarevich
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Petr I. Nikitin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
12
|
Belyaev IB, Zelepukin IV, Kotelnikova PA, Tikhonowski GV, Popov AA, Kapitannikova AY, Barman J, Kopylov AN, Bratashov DN, Prikhozhdenko ES, Kabashin AV, Deyev SM, Zvyagin AV. Laser-Synthesized Germanium Nanoparticles as Biodegradable Material for Near-Infrared Photoacoustic Imaging and Cancer Phototherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307060. [PMID: 38516744 PMCID: PMC11132077 DOI: 10.1002/advs.202307060] [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: 09/25/2023] [Revised: 02/20/2024] [Indexed: 03/23/2024]
Abstract
Biodegradable nanomaterials can significantly improve the safety profile of nanomedicine. Germanium nanoparticles (Ge NPs) with a safe biodegradation pathway are developed as efficient photothermal converters for biomedical applications. Ge NPs synthesized by femtosecond-laser ablation in liquids rapidly dissolve in physiological-like environment through the oxidation mechanism. The biodegradation of Ge nanoparticles is preserved in tumor cells in vitro and in normal tissues in mice with a half-life as short as 3.5 days. Biocompatibility of Ge NPs is confirmed in vivo by hematological, biochemical, and histological analyses. Strong optical absorption of Ge in the near-infrared spectral range enables photothermal treatment of engrafted tumors in vivo, following intravenous injection of Ge NPs. The photothermal therapy results in a 3.9-fold reduction of the EMT6/P adenocarcinoma tumor growth with significant prolongation of the mice survival. Excellent mass-extinction of Ge NPs (7.9 L g-1 cm-1 at 808 nm) enables photoacoustic imaging of bones and tumors, following intravenous and intratumoral administrations of the nanomaterial. As such, strongly absorbing near-infrared-light biodegradable Ge nanomaterial holds promise for advanced theranostics.
Collapse
Affiliation(s)
- Iaroslav B. Belyaev
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute)Moscow115409Russia
| | - Ivan V. Zelepukin
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
- Department of Medicinal ChemistryUppsala UniversityUppsala751 23Sweden
| | - Polina A. Kotelnikova
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
| | - Gleb V. Tikhonowski
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute)Moscow115409Russia
| | - Anton A. Popov
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute)Moscow115409Russia
| | | | - Jugal Barman
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
| | - Alexey N. Kopylov
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute)Moscow115409Russia
| | | | | | - Andrei V. Kabashin
- CNRSLP3Campus de Luminy – Case 917Aix Marseille UniversityMarseilleCedex13288France
| | - Sergey M. Deyev
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute)Moscow115409Russia
- Institute of Molecular TheranosticsSechenov UniversityMoscow119435Russia
| | - Andrei V. Zvyagin
- Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of SciencesMoscow117997Russia
- Institute of Molecular TheranosticsSechenov UniversityMoscow119435Russia
- MQ Photonics CentreMacquarie UniversitySydney2109Australia
| |
Collapse
|
13
|
Estapé Senti M, García Del Valle L, Schiffelers RM. mRNA delivery systems for cancer immunotherapy: Lipid nanoparticles and beyond. Adv Drug Deliv Rev 2024; 206:115190. [PMID: 38307296 DOI: 10.1016/j.addr.2024.115190] [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: 10/31/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
mRNA-based vaccines are emerging as a promising alternative to standard cancer treatments and the conventional vaccines. Moreover, the FDA-approval of three nucleic acid based therapeutics (Onpattro, BNT162b2 and mRNA-1273) has further increased the interest and trust on this type of therapeutics. In order to achieve a significant therapeutic efficacy, the mRNA needs from a drug delivery system. In the last years, several delivery platforms have been explored, being the lipid nanoparticles (LNPs) the most well characterized and studied. A better understanding on how mRNA-based therapeutics operate (both the mRNA itself and the drug delivery system) will help to further improve their efficacy and safety. In this review, we will provide an overview of what mRNA cancer vaccines are and their mode of action and we will highlight the advantages and challenges of the different delivery platforms that are under investigation.
Collapse
Affiliation(s)
- Mariona Estapé Senti
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Lucía García Del Valle
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Raymond M Schiffelers
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands.
| |
Collapse
|
14
|
Wang L, Quine S, Frickenstein AN, Lee M, Yang W, Sheth VM, Bourlon MD, He Y, Lyu S, Garcia-Contreras L, Zhao YD, Wilhelm S. Exploring and Analyzing the Systemic Delivery Barriers for Nanoparticles. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2308446. [PMID: 38828467 PMCID: PMC11142462 DOI: 10.1002/adfm.202308446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Indexed: 06/05/2024]
Abstract
Most nanomedicines require efficient in vivo delivery to elicit diagnostic and therapeutic effects. However, en route to their intended tissues, systemically administered nanoparticles often encounter delivery barriers. To describe these barriers, we propose the term "nanoparticle blood removal pathways" (NBRP), which summarizes the interactions between nanoparticles and the body's various cell-dependent and cell-independent blood clearance mechanisms. We reviewed nanoparticle design and biological modulation strategies to mitigate nanoparticle-NBRP interactions. As these interactions affect nanoparticle delivery, we studied the preclinical literature from 2011-2021 and analyzed nanoparticle blood circulation and organ biodistribution data. Our findings revealed that nanoparticle surface chemistry affected the in vivo behavior more than other nanoparticle design parameters. Combinatory biological-PEG surface modification improved the blood area under the curve by ~418%, with a decrease in liver accumulation of up to 47%. A greater understanding of nanoparticle-NBRP interactions and associated delivery trends will provide new nanoparticle design and biological modulation strategies for safer, more effective, and more efficient nanomedicines.
Collapse
Affiliation(s)
- Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Skyler Quine
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Alex N. Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Michael Lee
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Wen Yang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Vinit M. Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Margaret D. Bourlon
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yuxin He
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Shanxin Lyu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Lucila Garcia-Contreras
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yan D. Zhao
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73012, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), Norman, Oklahoma, 73019, USA
| |
Collapse
|
15
|
Zhao J, Zhang L, Li P, Liu S, Yu S, Chen Z, Zhu M, Xie S, Ling D, Li F. An Immunomodulatory Zinc-Alum/Ovalbumin Nanovaccine Boosts Cancer Metalloimmunotherapy Through Erythrocyte-Assisted Cascade Immune Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307389. [PMID: 38064201 PMCID: PMC10853754 DOI: 10.1002/advs.202307389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/29/2023] [Indexed: 02/10/2024]
Abstract
Cancer therapeutic vaccines are powerful tools for immune system activation and eliciting protective responses against tumors. However, their efficacy has often been hindered by weak and slow immune responses. Here, the authors introduce an immunization strategy employing senescent erythrocytes to facilitate the accumulation of immunomodulatory zinc-Alum/ovalbumin (ZAlum/OVA) nanovaccines within both the spleen and solid tumors by temporarily saturating liver macrophages. This approach sets the stage for boosted cancer metalloimmunotherapy through a cascade immune activation. The accumulation of ZAlum/OVA nanovaccines in the spleen substantially enhances autophagy-dependent antigen presentation in dendritic cells, rapidly initiating OVA-specific T-cell responses against solid tumors. Concurrently, ZAlum/OVA nanovaccines accumulated in the tumor microenvironment trigger immunogenic cell death, leading to the induction of individualized tumor-associated antigen-specific T cell responses and increased T cell infiltration. This erythrocyte-assisted cascade immune activation using ZAlum/OVA nanovaccines results in rapid and robust antitumor immunity induction, holding great potential for clinical cancer metalloimmunotherapy.
Collapse
Affiliation(s)
- Jing Zhao
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Lingxiao Zhang
- Interdisciplinary Nanoscience Center (iNANO)Aarhus UniversityAarhusC DK‐8000Denmark
| | - Pin Li
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Shanbiao Liu
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Shiyi Yu
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Zheng Chen
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Mingjian Zhu
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Shangzhi Xie
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Daishun Ling
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
- Frontiers Science Center for Transformative MoleculesSchool of Chemistry and Chemical EngineeringSchool of Biomedical EngineeringNational Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240P. R. China
- World Laureates Association (WLA) LaboratoriesShanghai201203P. R. China
| | - Fangyuan Li
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
- World Laureates Association (WLA) LaboratoriesShanghai201203P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhou310009P. R. China
| |
Collapse
|
16
|
Komedchikova EN, Kolesnikova OA, Syuy AV, Volkov VS, Deyev SM, Nikitin MP, Shipunova VO. Targosomes: Anti-HER2 PLGA nanocarriers for bioimaging, chemotherapy and local photothermal treatment of tumors and remote metastases. J Control Release 2024; 365:317-330. [PMID: 37996056 DOI: 10.1016/j.jconrel.2023.11.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023]
Abstract
Developing combined cancer therapy strategies is of utmost importance as it can enhance treatment efficacy, overcome drug resistance, and ultimately improve patient outcomes by targeting multiple pathways and mechanisms involved in cancer growth and progression. Specifically, the potential of developing a combination chemo&photothermal therapy using targeted polymer nanoparticles as nanocarriers offers a promising approach for synergistic cancer treatment by combining the benefits of both therapies, such as targeted drug delivery and localized hyperthermia. Here, we report the first targeted anti-HER2 PLGA nanocarriers, called targosomes, that simultaneously possess photothermal, chemotherapeutic and diagnostic properties using only molecular payloads. Biocompatible poly(lactic-co-glycolic acid), PLGA, nanoparticles were loaded with photosensitizer phthalocyanine, diagnostic dye Nile Blue, and chemotherapeutic drug irinotecan, which was chosen as a result of screening a panel of theragnostic nanoparticles. The targeted delivery to cell surface oncomarker HER2 was ensured by nanoparticle modification with the anti-HER2 monoclonal antibody, trastuzumab, using the one-pot synthesis method without chemical conjugation. The irradiation tests revealed prominent photothermal properties of nanoparticles, namely heating by 35 °C in 10 min. Nanoparticles exhibited a 7-fold increase in binding and nearly an 18-fold increase in cytotoxicity for HER2-overexpressing cells compared to cells lacking HER2 expression. This enhancement of cytotoxicity was further amplified by >20-fold under NIR light irradiation. In vivo studies proved the efficacy of nanoparticles for bioimaging of primary tumor and metastasis sites and demonstrated 93% tumor growth inhibition, making these nanoparticles excellent candidates for translation into theragnostic applications.
Collapse
Affiliation(s)
- E N Komedchikova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - O A Kolesnikova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - A V Syuy
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - V S Volkov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - S M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - M P Nikitin
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - V O Shipunova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia.
| |
Collapse
|
17
|
Liu C, Hern FY, Shakil A, Temburnikar K, Chambon P, Liptrott N, McDonald TO, Neary M, Flexner C, Owen A, Meyers CF, Rannard SP. Polymer-prodrug conjugates as candidates for degradable, long-acting implants, releasing the water-soluble nucleoside reverse-transcriptase inhibitor emtricitabine. J Mater Chem B 2023; 11:11532-11543. [PMID: 37955203 PMCID: PMC10718295 DOI: 10.1039/d3tb02268d] [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: 09/28/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
Circulating, soluble polymer-drug conjugates have been utilised for many years to aid the delivery of sensitive, poorly-soluble or cytotoxic drugs, prolong circulation times or minimise side effects. Long-acting therapeutics are increasing in their healthcare importance, with intramuscular and subcutaneous administration of liquid formulations being most common. Degradable implants also offer opportunities and the use of polymer-prodrug conjugates as implant materials has not been widely reported in this context. Here, the potential for polymer-prodrug conjugates of the water soluble nucleoside reverse transciption inhibitor emtricitabine (FTC) is studied. A novel diol monomer scaffold, allowing variation of prodrug substitution, has been used to form polyesters and polycarbonates by step-growth polymerisation. Materials have been screened for physical properties that enable implant formation, studied for drug release to provide mechanistic insights, and tunable prolonged release of FTC has been demonstrated over a period of at least two weeks under relevant physiological conditions.
Collapse
Affiliation(s)
- Chung Liu
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Faye Y Hern
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Anika Shakil
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Kartik Temburnikar
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Pierre Chambon
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Neill Liptrott
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L7 3NY, UK
| | - Tom O McDonald
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| | - Megan Neary
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L7 3NY, UK
| | - Charles Flexner
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Andrew Owen
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L7 3NY, UK
| | - Caren Freel Meyers
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Steve P Rannard
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool, L69 7ZD, UK.
- Materials Innovation Factory, University of Liverpool, Crown Street, L69 7ZD, UK
- Centre of Excellence in Long-acting Therapeutics (CELT), University of Liverpool, Liverpool, L7 3NY, UK
| |
Collapse
|
18
|
Qin M, Xia H, Xu W, Chen B, Wang Y. The spatiotemporal journey of nanomedicines in solid tumors on their therapeutic efficacy. Adv Drug Deliv Rev 2023; 203:115137. [PMID: 37949414 DOI: 10.1016/j.addr.2023.115137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
The rapid development of nanomedicines is revolutionizing the landscape of cancer treatment, while effectively delivering them into solid tumors remains a formidable challenge. Currently, there is a huge disconnect on therapeutic response between regulatory approved nanomedicines and laboratory reported nanoparticles. The discrepancy is mainly resulted from the failure of using the classic overall pharmacokinetics behaviors of nanomedicines in tumors to predict the antitumor efficacy. Increasing evidence has revealed that the therapeutic efficacy predominantly relies on the intratumoral spatiotemporal distribution of nanomedicines. This review focuses on the spatiotemporal distribution of systemically administered chemotherapeutic nanomedicines in solid tumor. Firstly, the intratumoral biological barriers that regulate the spatiotemporal distribution of nanomedicines are described in detail. Next, the influences on antitumor efficacy caused by the spatial distribution and temporal drug release of nanomedicines are emphatically analyzed. Then, current methodologies for evaluating the spatiotemporal distribution of nanomedicines are summarized. Finally, the advanced strategies to positively modulate the spatiotemporal distribution of nanomedicines for an optimal tumor therapy are comprehensively reviewed.
Collapse
Affiliation(s)
- Mengmeng Qin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Heming Xia
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wenhao Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China; Chemical Biology Center, Peking University, Beijing, China.
| |
Collapse
|
19
|
Ivanova A, Badertscher L, O'Driscoll G, Bergman J, Gordon E, Gunnarsson A, Johansson C, Munson MJ, Spinelli C, Torstensson S, Vilén L, Voirel A, Wiseman J, Rak J, Dekker N, Lázaro‐Ibáñez E. Creating Designer Engineered Extracellular Vesicles for Diverse Ligand Display, Target Recognition, and Controlled Protein Loading and Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304389. [PMID: 37867228 PMCID: PMC10700174 DOI: 10.1002/advs.202304389] [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: 06/30/2023] [Revised: 09/19/2023] [Indexed: 10/24/2023]
Abstract
Efficient and targeted delivery of therapeutic agents remains a bottleneck in modern medicine. Here, biochemical engineering approaches to advance the repurposing of extracellular vesicles (EVs) as drug delivery vehicles are explored. Targeting ligands such as the sugar GalNAc are displayed on the surface of EVs using a HaloTag-fused to a protein anchor that is enriched on engineered EVs. These EVs are successfully targeted to human primary hepatocytes. In addition, the authors are able to decorate EVs with an antibody that recognizes a GLP1 cell surface receptor by using an Fc and Fab region binding moiety fused to an anchor protein, and they show that this improves EV targeting to cells that overexpress the receptor. The authors also use two different protein-engineering approaches to improve the loading of Cre recombinase into the EV lumen and demonstrate that functional Cre protein is delivered into cells in the presence of chloroquine, an endosomal escape enhancer. Lastly, engineered EVs are well tolerated upon intravenous injection into mice without detectable signs of liver toxicity. Collectively, the data show that EVs can be engineered to improve cargo loading and specific cell targeting, which will aid their transformation into tailored drug delivery vehicles.
Collapse
Affiliation(s)
- Alena Ivanova
- Discovery BiologyDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Lukas Badertscher
- Translational GenomicsDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
- Present address:
Myllia Biotechnology GmbHAm Kanal 27Vienna1110Austria
| | - Gwen O'Driscoll
- Discovery BiologyDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
- Advanced Drug DeliveryPharmaceutical SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
- Present address:
Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUK
| | - Joakim Bergman
- Medicinal ChemistryResearch and Early Development Cardiovascular, Renal and MetabolismBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Euan Gordon
- Discovery BiologyDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Anders Gunnarsson
- Structure and BiophysicsDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Camilla Johansson
- Clinical Pharmacology and Safety SciencesSweden Imaging HubBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Michael J. Munson
- Advanced Drug DeliveryPharmaceutical SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Cristiana Spinelli
- Research Institute of the McGill University Health CentreGlen SiteMcGill UniversityMontrealQuebecH4A 3J1Canada
| | - Sara Torstensson
- Translational GenomicsDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Liisa Vilén
- DMPKResearch and Early Development Cardiovascular, Renal and MetabolismBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Andrei Voirel
- Medicinal ChemistryResearch and Early Development Cardiovascular, Renal and MetabolismBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - John Wiseman
- Translational GenomicsDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Janusz Rak
- Research Institute of the McGill University Health CentreGlen SiteMcGill UniversityMontrealQuebecH4A 3J1Canada
| | - Niek Dekker
- Discovery BiologyDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Elisa Lázaro‐Ibáñez
- Discovery BiologyDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
- Advanced Drug DeliveryPharmaceutical SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| |
Collapse
|
20
|
Guo L, Zhao Q, Zheng LW, Wang M. Multifunctional Nanofibrous Scaffolds Capable of Localized Delivery of Theranostic Nanoparticles for Postoperative Cancer Management. Adv Healthc Mater 2023; 12:e2302484. [PMID: 37702133 DOI: 10.1002/adhm.202302484] [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: 08/01/2023] [Indexed: 09/14/2023]
Abstract
Postoperative recovery of cancer patients can be affected by complications, such as tissue dysfunction or disability caused by tissue resection, and also cancer recurrence resulting from residual cancer cells. Despite impressive progress made for tissue engineering scaffolds that assist tissue regeneration for postoperative cancer patients, the majority of existing tissue engineering scaffolds still lack functions for monitoring and killing residual cancer cells, if there are any, upon their detection. In this study, multifunctional scaffolds that comprise biodegradable nanofibers and core-shell structured microspheres encapsulated with theranostic nanoparticles (NPs) are developed. The multifunctional scaffolds possess an extracellular matrix-like nanofibrous architecture and soft tissue-like mechanical properties, making them excellent tissue engineering patch candidates for assisting in the repair and regeneration of tissues at the cancerous sites after surgery. Furthermore, they are capable of localized delivery of theranostic NPs upon quick degradation of core-shell structured microspheres that contain theranostic NPs. Leveraging on folic acid-mediated ligand-receptor binding, surface-enhanced Raman scattering activity, and near-infrared-responsive photothermal effect of the theranostic gold NPs (AuNPs) delivered locally, the multifunctional scaffolds display excellent active targeting, diagnosis, and photothermal therapy functions for cancer cells, showing great promise for adaptive postoperative cancer management.
Collapse
Affiliation(s)
- Lin Guo
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong
| | - Qilong Zhao
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong
- Institute of Biomedical & Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
| | - Li-Wu Zheng
- Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong
| | - Min Wang
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong
| |
Collapse
|
21
|
Belyaev IB, Zelepukin IV, Tishchenko VK, Petriev VM, Trushina DB, Klimentov SM, Zavestovskaya IN, Ivanov SA, Kaprin AD, Deyev SM, Kabashin AV. Nanoparticles based on MIL-101 metal-organic frameworks as efficient carriers of therapeutic 188Re radionuclide for nuclear medicine. NANOTECHNOLOGY 2023; 35:075103. [PMID: 37963406 DOI: 10.1088/1361-6528/ad0c74] [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: 08/28/2023] [Accepted: 11/14/2023] [Indexed: 11/16/2023]
Abstract
Nuclear medicine presents one of the most promising modalities for efficient non-invasive treatment of a variety of cancers, but the application of radionuclides in cancer therapy and diagnostics is severely limited by their nonspecific tissue accumulation and poor biocompatibility. Here, we explore the use of nanosized metal-organic frameworks (MOFs) as carriers of radionuclides to order to improve their delivery to tumour. To demonstrate the concept, we prepared polymer-coated MIL-101(Cr)-NH2MOFs and conjugated them with clinically utilized radionuclide188Re. The nanoparticles demonstrated high loading efficacy of radionuclide reaching specific activity of 49 MBq mg-1. Pharmacokinetics of loaded MOFs was investigated in mice bearing colon adenocarcinoma. The biological half-life of the radionuclide in blood was (20.9 ± 1.3) h, and nanoparticles enabled it to passively accumulate and retain in the tumour. The radionuclide delivery with MOFs led to a significant decrease of radioactivity uptake by the thyroid gland and stomach as compared with perrhenate salt injection, which is beneficial for reducing the side toxicity of nuclear therapy. The reported data on the functionalization and pharmacokinetics of MIL-101(Cr)-NH2for radionuclide delivery unveils the promising potential of these MOFs for nuclear medicine.
Collapse
Affiliation(s)
- Iaroslav B Belyaev
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Ivan V Zelepukin
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Victoria K Tishchenko
- A. Tsyb Medical Radiological Research Centre, Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia
| | - Vasiliy M Petriev
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
- A. Tsyb Medical Radiological Research Centre, Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia
| | - Daria B Trushina
- Federal Research Center 'Crystallography and Photonics', Russian Academy of Sciences, Moscow 119333, Russia
- Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Sergey M Klimentov
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
| | - Irina N Zavestovskaya
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
| | - Sergey A Ivanov
- A. Tsyb Medical Radiological Research Centre, Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia
| | - Andrey D Kaprin
- A. Tsyb Medical Radiological Research Centre, Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia
| | - Sergey M Deyev
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
- Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
- Kazan Federal University, Institute of Fundamental Medicine and Biology, 420008, Kazan, Russia
| | - Andrei V Kabashin
- Aix Marseille University, CNRS, LP3, Campus de Luminy, Case 917, F-13288, Marseille, France
| |
Collapse
|
22
|
Granata S, Stallone G, Zaza G. mRNA as a medicine in nephrology: the future is now. Clin Kidney J 2023; 16:2349-2356. [PMID: 38046026 PMCID: PMC10689145 DOI: 10.1093/ckj/sfad196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Indexed: 12/05/2023] Open
Abstract
The successful employment of messenger RNA (mRNA) as vaccine therapy for the prevention of COVID-19 infection has spotlighted the attention of scientific community onto the potential clinical application of these molecules as innovative and alternative therapeutic approaches in different fields of medicine. As therapy, mRNAs may be advantageous due to their unique biological properties of targeting almost any genetic component within the cell, many of which may be unreachable using other pharmacological/therapeutic approaches, and encoding any proteins and peptides without the need for their transport into the nuclei of the target cells. Additionally, these molecules may be rapidly designed/produced and clinically tested. Once the chemistry of the RNA and its delivery system are optimized, the cost of developing novel variants of these medications for new selected clinical disorders is significantly reduced. However, although potentially useful as new therapeutic weapons against several kidney diseases, the complex architecture of kidney and the inability of nanoparticles that accommodate oligonucleotides to cross the integral glomerular filtration barrier have largely decreased their potential employment in nephrology. However, in the next few years, the technical improvements in mRNA that increase translational efficiency, modulate innate and adaptive immunogenicity, and increase their delivery at the site of action will overcome these limitations. Therefore, this review has the scope of summarizing the key strengths of these RNA-based therapies and illustrating potential future directions and challenges of this promising technology for widespread therapeutic use in nephrology.
Collapse
Affiliation(s)
- Simona Granata
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gianluigi Zaza
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
23
|
Hasan N, Imran M, Jain D, Jha SK, Nadaf A, Chaudhary A, Rafiya K, Jha LA, Almalki WH, Mohammed Y, Kesharwani P, Ahmad FJ. Advanced targeted drug delivery by bioengineered white blood cell-membrane camouflaged nanoparticulate delivery nanostructures. ENVIRONMENTAL RESEARCH 2023; 238:117007. [PMID: 37689337 DOI: 10.1016/j.envres.2023.117007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/22/2023] [Accepted: 08/26/2023] [Indexed: 09/11/2023]
Abstract
Targeted drug delivery has emerged as a pivotal approach within precision medicine, aiming to optimize therapeutic efficacy while minimizing systemic side effects. Leukocyte membrane coated nanoparticles (NPs) have attracted a lot of interest as an effective approach for delivering targeted drugs, capitalizing on the natural attributes of leukocytes to achieve site-specific accumulation, and heightened therapeutic outcomes. An overview of the present state of the targeted medication delivery research is given in this review. Notably, Leukocyte membrane-coated NPs offer inherent advantages such as immune evasion, extended circulation half-life, and precise homing to inflamed or diseased tissues through specific interactions with adhesion molecules. leukocyte membrane-coated NPs hold significant promise in advancing targeted drug delivery for precision medicine. As research progresses, they are anticipated to contribute to improved therapeutic outcomes, enabling personalized and effective treatments for a wide range of diseases and conditions. The review covers the method of preparation, characterization, and biological applications of leucocytic membrane coated NPs. Further, patents related factors, gap of translation from laboratory to clinic, and future prospective were discussed in detail. Overall, the review covers extensive literature to establish leucocytic membrane NPs for targeted drug delivery.
Collapse
Affiliation(s)
- Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammad Imran
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Dhara Jain
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Saurav Kumar Jha
- Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur, 208016, Uttar Pradesh, India
| | - Arif Nadaf
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Arshi Chaudhary
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Km Rafiya
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Laxmi Akhileshwar Jha
- H. K. College of Pharmacy, Mumbai University, Pratiksha Nagar, Jogeshwari, West Mumbai, 400102, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 24381, Saudi Arabia
| | - Yousuf Mohammed
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
24
|
Sell M, Lopes AR, Escudeiro M, Esteves B, Monteiro AR, Trindade T, Cruz-Lopes L. Application of Nanoparticles in Cancer Treatment: A Concise Review. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2887. [PMID: 37947732 PMCID: PMC10650201 DOI: 10.3390/nano13212887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023]
Abstract
Timely diagnosis and appropriate antitumoral treatments remain of utmost importance, since cancer remains a leading cause of death worldwide. Within this context, nanotechnology offers specific benefits in terms of cancer therapy by reducing its adverse effects and guiding drugs to selectively target cancer cells. In this comprehensive review, we have summarized the most relevant novel outcomes in the range of 2010-2023, covering the design and application of nanosystems for cancer therapy. We have established the general requirements for nanoparticles to be used in drug delivery and strategies for their uptake in tumor microenvironment and vasculature, including the reticuloendothelial system uptake and surface functionalization with protein corona. After a brief review of the classes of nanovectors, we have covered different classes of nanoparticles used in cancer therapies. First, the advances in the encapsulation of drugs (such as paclitaxel and fisetin) into nanoliposomes and nanoemulsions are described, as well as their relevance in current clinical trials. Then, polymeric nanoparticles are presented, namely the ones comprising poly lactic-co-glycolic acid, polyethylene glycol (and PEG dilemma) and dendrimers. The relevance of quantum dots in bioimaging is also covered, namely the systems with zinc sulfide and indium phosphide. Afterwards, we have reviewed gold nanoparticles (spheres and anisotropic) and their application in plasmon-induced photothermal therapy. The clinical relevance of iron oxide nanoparticles, such as magnetite and maghemite, has been analyzed in different fields, namely for magnetic resonance imaging, immunotherapy, hyperthermia, and drug delivery. Lastly, we have covered the recent advances in the systems using carbon nanomaterials, namely graphene oxide, carbon nanotubes, fullerenes, and carbon dots. Finally, we have compared the strategies of passive and active targeting of nanoparticles and their relevance in cancer theranostics. This review aims to be a (nano)mark on the ongoing journey towards realizing the remarkable potential of different nanoparticles in the realm of cancer therapeutics.
Collapse
Affiliation(s)
- Mariana Sell
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
| | - Ana Rita Lopes
- Faculty of Dental Medicine, Portuguese Catholic University, 3504-505 Viseu, Portugal;
| | - Maria Escudeiro
- Abel Salazar Biomedical Institute, University of Porto, 4050-313 Porto, Portugal;
| | - Bruno Esteves
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
- Centre for Natural Resources, Environment and Society-CERNAS-IPV Research Centre, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal
| | - Ana R. Monteiro
- Centro de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain;
| | - Tito Trindade
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Luísa Cruz-Lopes
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
- Centre for Natural Resources, Environment and Society-CERNAS-IPV Research Centre, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal
| |
Collapse
|
25
|
Gabashvili AN, Chmelyuk NS, Oda VV, Leonova MK, Sarkisova VA, Lazareva PA, Semkina AS, Belyakov NA, Nizamov TR, Nikitin PI. Magnetic and Fluorescent Dual-Labeled Genetically Encoded Targeted Nanoparticles for Malignant Glioma Cell Tracking and Drug Delivery. Pharmaceutics 2023; 15:2422. [PMID: 37896182 PMCID: PMC10609955 DOI: 10.3390/pharmaceutics15102422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Human glioblastoma multiforme (GBM) is a primary malignant brain tumor, a radically incurable disease characterized by rapid growth resistance to classical therapies, with a median patient survival of about 15 months. For decades, a plethora of approaches have been developed to make GBM therapy more precise and improve the diagnosis of this pathology. Targeted delivery mediated by the use of various molecules (monoclonal antibodies, ligands to overexpressed tumor receptors) is one of the promising methods to achieve this goal. Here we present a novel genetically encoded nanoscale dual-labeled system based on Quasibacillus thermotolerans (Qt) encapsulins exploiting biologically inspired designs with iron-containing nanoparticles as a cargo, conjugated with human fluorescent labeled transferrin (Tf) acting as a vector. It is known that the expression of transferrin receptors (TfR) in glioma cells is significantly higher compared to non-tumor cells, which enables the targeting of the resulting nanocarrier. The selectivity of binding of the obtained nanosystem to glioma cells was studied by qualitative and quantitative assessment of the accumulation of intracellular iron, as well as by magnetic particle quantification method and laser scanning confocal microscopy. Used approaches unambiguously demonstrated that transferrin-conjugated encapsulins were captured by glioma cells much more efficiently than by benign cells. The resulting bioinspired nanoplatform can be supplemented with a chemotherapeutic drug or genotherapeutic agent and used for targeted delivery of a therapeutic agent to malignant glioma cells. Additionally, the observed cell-assisted biosynthesis of magnetic nanoparticles could be an attractive way to achieve a narrow size distribution of particles for various applications.
Collapse
Affiliation(s)
- Anna N. Gabashvili
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov Street, 119991 Moscow, Russia; (A.N.G.)
| | - Nelly S. Chmelyuk
- Laboratory “Biomedical Nanomaterials”, National University of Science and Technology “MISIS”, Leninskiy Prospekt 4, 119049 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 1 Ostrovityanova Street, 117997 Moscow, Russia; (P.A.L.)
| | - Vera V. Oda
- MILLAB Group Ltd., 100/2 Dmitrovskoe Highway, 127247 Moscow, Russia
| | - Maria K. Leonova
- Department of Physical Chemistry, National University of Science and Technology “MISIS”, Leninskiy Prospekt 4, 119049 Moscow, Russia
| | - Viktoria A. Sarkisova
- Biology Faculty, Lomonosov Moscow State University, 1 Leninskiy Gory, 119234 Moscow, Russia
- Cell Proliferation Laboratory, Engelhardt Institute of Molecular Biology RAS, 32 Vavilov Street, 119991 Moscow, Russia
| | - Polina A. Lazareva
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 1 Ostrovityanova Street, 117997 Moscow, Russia; (P.A.L.)
| | - Alevtina S. Semkina
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 1 Ostrovityanova Street, 117997 Moscow, Russia; (P.A.L.)
- Department of Basic and Applied Neurobiology, Serbsky National Medical Research Center for Psychiatry and Narcology, 23 Kropotkinskiy Lane, 119991 Moscow, Russia
| | - Nikolai A. Belyakov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov Street, 119991 Moscow, Russia; (A.N.G.)
| | - Timur R. Nizamov
- Laboratory “Biomedical Nanomaterials”, National University of Science and Technology “MISIS”, Leninskiy Prospekt 4, 119049 Moscow, Russia
| | - Petr I. Nikitin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov Street, 119991 Moscow, Russia; (A.N.G.)
| |
Collapse
|
26
|
Lenders V, Koutsoumpou X, Phan P, Soenen SJ, Allegaert K, de Vleeschouwer S, Toelen J, Zhao Z, Manshian BB. Modulation of engineered nanomaterial interactions with organ barriers for enhanced drug transport. Chem Soc Rev 2023; 52:4672-4724. [PMID: 37338993 DOI: 10.1039/d1cs00574j] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
The biomedical use of nanoparticles (NPs) has been the focus of intense research for over a decade. As most NPs are explored as carriers to alter the biodistribution, pharmacokinetics and bioavailability of associated drugs, the delivery of these NPs to the tissues of interest remains an important topic. To date, the majority of NP delivery studies have used tumor models as their tool of interest, and the limitations concerning tumor targeting of systemically administered NPs have been well studied. In recent years, the focus has also shifted to other organs, each presenting their own unique delivery challenges to overcome. In this review, we discuss the recent advances in leveraging NPs to overcome four major biological barriers including the lung mucus, the gastrointestinal mucus, the placental barrier, and the blood-brain barrier. We define the specific properties of these biological barriers, discuss the challenges related to NP transport across them, and provide an overview of recent advances in the field. We discuss the strengths and shortcomings of different strategies to facilitate NP transport across the barriers and highlight some key findings that can stimulate further advances in this field.
Collapse
Affiliation(s)
- Vincent Lenders
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| | - Xanthippi Koutsoumpou
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| | - Philana Phan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Stefaan J Soenen
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Karel Allegaert
- Department of Hospital Pharmacy, Erasmus MC University Medical Center, CN Rotterdam, 3015, The Netherlands
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B3000 Leuven, Belgium
- Leuven Child and Youth Institute, KU Leuven, 3000 Leuven, Belgium
- Woman and Child, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Steven de Vleeschouwer
- Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Jaan Toelen
- Leuven Child and Youth Institute, KU Leuven, 3000 Leuven, Belgium
- Woman and Child, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| |
Collapse
|
27
|
Wang H, Lin S, Wu X, Jiang K, Lu H, Zhan C. Interplay between Liposomes and IgM: Principles, Challenges, and Opportunities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301777. [PMID: 37150860 PMCID: PMC10369250 DOI: 10.1002/advs.202301777] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/14/2023] [Indexed: 05/09/2023]
Abstract
Liposomes have received tremendous attention as a class of versatile pharmaceutical vehicles of great potential over the past several decades. However, the application of liposomes encounters major challenges due to the knowledge gaps in their in vivo delivery process. Immunoglobulin M (IgM) displays both pervasiveness and complexity in regulating the biological functions as well as eliciting adverse effects of liposomes. Understanding, mitigating, and exploiting the duality of IgM are prerequisites for achieving various biomedical applications of liposomes. In this review, the intricate relationship between liposomes and their biological environments has been summarized, with an emphasis on the regulatory effects of IgM on in vivo performance of liposomes. Corresponding solutions have also been discussed to evade IgM-mediated opsonization for safe and efficient drug delivery.
Collapse
Affiliation(s)
- Huan Wang
- School of PharmacyNaval Medical UniversityShanghai200433P. R. China
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of PharmacologySchool of Basic Medical SciencesFudan UniversityShanghai201399P. R. China
| | - Shiqi Lin
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of PharmacologySchool of Basic Medical SciencesFudan UniversityShanghai201399P. R. China
| | - Xiying Wu
- Shanghai Skin Disease HospitalTongji University School of MedicineShanghai200443China
| | - Kuan Jiang
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of PharmacologySchool of Basic Medical SciencesFudan UniversityShanghai201399P. R. China
| | - Huiping Lu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of PharmacologySchool of Basic Medical SciencesFudan UniversityShanghai201399P. R. China
| | - Changyou Zhan
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of PharmacologySchool of Basic Medical SciencesFudan UniversityShanghai201399P. R. China
- Shanghai Engineering Research Center for Synthetic ImmunologyFudan UniversityShanghai200032P. R. China
| |
Collapse
|
28
|
Mochalova EN, Egorova EA, Komarova KS, Shipunova VO, Khabibullina NF, Nikitin PI, Nikitin MP. Comparative Study of Nanoparticle Blood Circulation after Forced Clearance of Own Erythrocytes (Mononuclear Phagocyte System-Cytoblockade) or Administration of Cytotoxic Doxorubicin- or Clodronate-Loaded Liposomes. Int J Mol Sci 2023; 24:10623. [PMID: 37445804 DOI: 10.3390/ijms241310623] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/05/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Recent developments in the field of nanomedicine have introduced a wide variety of nanomaterials that are capable of recognizing and killing tumor cells with increased specificity. A major limitation preventing the widespread introduction of nanomaterials into the clinical setting is their fast clearance from the bloodstream via the mononuclear phagocyte system (MPS). One of the most promising methods used to overcome this limitation is the MPS-cytoblockade, which forces the MPS to intensify the clearance of erythrocytes by injecting allogeneic anti-erythrocyte antibodies and, thus, significantly prolongs the circulation of nanoagents in the blood. However, on the way to the clinical application of this approach, the question arises whether the induced suppression of macrophage phagocytosis via the MPS-cytoblockade could pose health risks. Here, we show that highly cytotoxic doxorubicin- or clodronate-loaded liposomes, which are widely used for cancer therapy and biomedical research, induce a similar increase in the nanoparticle blood circulation half-life in mice as the MPS-cytoblockade, which only gently and temporarily saturates the macrophages with the organism's own erythrocytes. This result suggests that from the point of view of in vivo macrophage suppression, the MPS-cytoblockade should be less detrimental than the liposomal anti-cancer drugs that are already approved for clinical application while allowing for the substantial improvement in the nanoagent effectiveness.
Collapse
Affiliation(s)
- Elizaveta N Mochalova
- Nanobiomedicine Division, Sirius University of Science and Technology, 1 Olimpiyskiy Ave, 354340 Sirius, Russia
- Moscow Institute of Physics and Technology, 1A Kerchenskaya St., 117303 Moscow, Russia
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia
| | - Elena A Egorova
- Nanobiomedicine Division, Sirius University of Science and Technology, 1 Olimpiyskiy Ave, 354340 Sirius, Russia
| | - Kristina S Komarova
- Moscow Institute of Physics and Technology, 1A Kerchenskaya St., 117303 Moscow, Russia
| | - Victoria O Shipunova
- Nanobiomedicine Division, Sirius University of Science and Technology, 1 Olimpiyskiy Ave, 354340 Sirius, Russia
- Moscow Institute of Physics and Technology, 1A Kerchenskaya St., 117303 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia
| | - Nelli F Khabibullina
- Moscow Institute of Physics and Technology, 1A Kerchenskaya St., 117303 Moscow, Russia
| | - Petr I Nikitin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia
| | - Maxim P Nikitin
- Nanobiomedicine Division, Sirius University of Science and Technology, 1 Olimpiyskiy Ave, 354340 Sirius, Russia
- Moscow Institute of Physics and Technology, 1A Kerchenskaya St., 117303 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia
| |
Collapse
|
29
|
Wen P, Ke W, Dirisala A, Toh K, Tanaka M, Li J. Stealth and pseudo-stealth nanocarriers. Adv Drug Deliv Rev 2023; 198:114895. [PMID: 37211278 DOI: 10.1016/j.addr.2023.114895] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
The stealth effect plays a central role on capacitating nanomaterials for drug delivery applications through improving the pharmacokinetics such as blood circulation, biodistribution, and tissue targeting. Here based on a practical analysis of stealth efficiency and a theoretical discussion of relevant factors, we provide an integrated material and biological perspective in terms of engineering stealth nanomaterials. The analysis surprisingly shows that more than 85% of the reported stealth nanomaterials encounter a rapid drop of blood concentration to half of the administered dose within 1 h post administration although a relatively long β-phase is observed. A term, pseudo-stealth effect, is used to delineate this common pharmacokinetics behavior of nanomaterials, that is, dose-dependent nonlinear pharmacokinetics because of saturating or depressing bio-clearance of RES. We further propose structural holism can be a watershed to improve the stealth effect; that is, the whole surface structure and geometry play important roles, rather than solely relying on a single factor such as maximizing repulsion force through polymer-based steric stabilization (e.g., PEGylation) or inhibiting immune attack through a bio-inspired component. Consequently, engineering delicate structural hierarchies to minimize attractive binding sites, that is, minimal charges/dipole and hydrophobic domain, becomes crucial. In parallel, the pragmatic implementation of the pseudo-stealth effect and dynamic modulation of the stealth effect are discussed for future development.
Collapse
Affiliation(s)
- Panyue Wen
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Wendong Ke
- Chemical Macromolecule Division, Asymchem Life Science (Tianjin) Co., Ltd. No. 71, Seventh Avenue, TEDA Tianjin 300457, P.R. China
| | - Anjaneyulu Dirisala
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazuko Toh
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Masaru Tanaka
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Junjie Li
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
| |
Collapse
|
30
|
Li Z, Zhu Y, Zeng H, Wang C, Xu C, Wang Q, Wang H, Li S, Chen J, Xiao C, Yang X, Li Z. Mechano-boosting nanomedicine antitumour efficacy by blocking the reticuloendothelial system with stiff nanogels. Nat Commun 2023; 14:1437. [PMID: 36918575 PMCID: PMC10015032 DOI: 10.1038/s41467-023-37150-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
Nanomedicine has been developed for cancer therapy over several decades, while rapid clearance from blood circulation by reticuloendothelial system (RES) severely limits nanomedicine antitumour efficacy. We design a series of nanogels with distinctive stiffness and investigate how nanogel mechanical properties could be leveraged to overcome RES. Stiff nanogels are injected preferentially to abrogate uptake capacity of macrophages and temporarily block RES, relying on inhibition of clathrin and prolonged liver retention. Afterwards, soft nanogels deliver doxorubicin (DOX) with excellent efficiency, reflected in high tumour accumulation, deep tumour penetration and outstanding antitumour efficacy. In this work, we combine the advantage of stiff nanogels in RES-blockade with the superiority of soft nanogels in drug delivery leads to the optimum tumour inhibition effect, which is defined as mechano-boosting antitumour strategy. Clinical implications of stiffness-dependent RES-blockade are also confirmed by promoting antitumour efficacy of commercialized nanomedicines, such as Doxil and Abraxane.
Collapse
Affiliation(s)
- Zheng Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Yabo Zhu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Haowen Zeng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Chong Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Chen Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Qiang Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Huimin Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Shiyou Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Jitang Chen
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Chen Xiao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China
- GBA Research Innovation Institute for Nanotechnology, 510530, Guangzhou, Guangdong, P. R. China
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China.
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China.
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, 430074, Wuhan, P. R. China.
| |
Collapse
|
31
|
Targeted Two-Step Delivery of Oncotheranostic Nano-PLGA for HER2-Positive Tumor Imaging and Therapy In Vivo: Improved Effectiveness Compared to One-Step Strategy. Pharmaceutics 2023; 15:pharmaceutics15030833. [PMID: 36986694 PMCID: PMC10053351 DOI: 10.3390/pharmaceutics15030833] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Therapy for aggressive metastatic breast cancer remains a great challenge for modern biomedicine. Biocompatible polymer nanoparticles have been successfully used in clinic and are seen as a potential solution. Specifically, researchers are exploring the development of chemotherapeutic nanoagents targeting the membrane-associated receptors of cancer cells, such as HER2. However, there are no targeting nanomedications that have been approved for human cancer therapy. Novel strategies are being developed to alter the architecture of agents and optimize their systemic administration. Here, we describe a combination of these approaches, namely, the design of a targeted polymer nanocarrier and a method for its systemic delivery to the tumor site. Namely, PLGA nanocapsules loaded with a diagnostic dye, Nile Blue, and a chemotherapeutic compound, doxorubicin, are used for two-step targeted delivery using the concept of tumor pre-targeting through the barnase/barstar protein “bacterial superglue”. The first pre-targeting component consists of an anti-HER2 scaffold protein, DARPin9_29 fused with barstar, Bs-DARPin9_29, and the second component comprises chemotherapeutic PLGA nanocapsules conjugated to barnase, PLGA-Bn. The efficacy of this system was evaluated in vivo. To this aim, we developed an immunocompetent BALB/c mouse tumor model with a stable expression of human HER2 oncomarkers to test the potential of two-step delivery of oncotheranostic nano-PLGA. In vitro and ex vivo studies confirmed HER2 receptor stable expression in the tumor, making it a feasible tool for HER2-targeted drug evaluation. We demonstrated that two-step delivery was more effective than one-step delivery for both imaging and tumor therapy: two-step delivery had higher imaging capabilities than one-step and a tumor growth inhibition of 94.9% in comparison to 68.4% for the one-step strategy. The barnase*barstar protein pair has been proven to possess excellent biocompatibility, as evidenced by the successful completion of biosafety tests assessing immunogenicity and hemotoxicity. This renders the protein pair a highly versatile tool for pre-targeting tumors with various molecular profiles, thereby enabling the development of personalized medicine.
Collapse
|
32
|
Lu J, Gao X, Wang S, He Y, Ma X, Zhang T, Liu X. Advanced strategies to evade the mononuclear phagocyte system clearance of nanomaterials. EXPLORATION (BEIJING, CHINA) 2023; 3:20220045. [PMID: 37323617 PMCID: PMC10191055 DOI: 10.1002/exp.20220045] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/12/2022] [Indexed: 06/17/2023]
Abstract
Nanomaterials are promising carriers to improve the bioavailability and therapeutic efficiency of drugs by providing preferential drug accumulation at their sites of action, but their delivery efficacy is severely limited by a series of biological barriers, especially the mononuclear phagocytic system (MPS)-the first and major barrier encountered by systemically administered nanomaterials. Herein, the current strategies for evading the MPS clearance of nanomaterials are summarized. First, engineering nanomaterials methods including surface modification, cell hitchhiking, and physiological environment modulation to reduce the MPS clearance are explored. Second, MPS disabling methods including MPS blockade, suppression of macrophage phagocytosis, and macrophages depletion are examined. Last, challenges and opportunities in this field are further discussed.
Collapse
Affiliation(s)
- Junjie Lu
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of EducationCollege of Chemistry and Materials ScienceNorthwest UniversityXi'anChina
| | - Xiao Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China of the Ministry of EducationSchool of MedicineNorthwest UniversityXi'anChina
| | - Siyao Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China of the Ministry of EducationSchool of MedicineNorthwest UniversityXi'anChina
| | - Yuan He
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of EducationCollege of Chemistry and Materials ScienceNorthwest UniversityXi'anChina
| | - Xiaowei Ma
- National Center for Veterinary Drug Safety EvaluationCollege of Veterinary MedicineChina Agricultural UniversityBeijingChina
| | - Tingbin Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of EducationCollege of Chemistry and Materials ScienceNorthwest UniversityXi'anChina
| | - Xiaoli Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China of the Ministry of EducationSchool of MedicineNorthwest UniversityXi'anChina
- Institute of Regenerative and Reconstructive MedicineMed‐X InstituteNational Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
33
|
Tumor-Targeted Erythrocyte Membrane Nanoparticles for Theranostics of Triple-Negative Breast Cancer. Pharmaceutics 2023; 15:pharmaceutics15020350. [PMID: 36839675 PMCID: PMC9966336 DOI: 10.3390/pharmaceutics15020350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 01/24/2023] Open
Abstract
Triple-negative breast cancer (TNBC) cells do not contain various receptors for targeted treatment, a reason behind the poor prognosis of this disease. In this study, biocompatible theranostic erythrocyte-derived nanoparticles (EDNs) were developed and evaluated for effective early diagnosis and treatment of TNBC. The anti-cancer drug, doxorubicin (DOX), was encapsulated into the EDNs and diagnostic quantum dots (QDs) were incorporated into the lipid bilayers of EDNs for tumor bio-imaging. Then, anti-epidermal growth factor receptor (EGFR) antibody molecules were conjugated to the surface of EDNs for TNBC targeting (iEDNs). According to the confocal microscopic analyses and biodistribution assay, iEDNs showed a higher accumulation in EGFR-positive MDA-MB-231 cancers in vitro as well as in vivo, compared to untargeted EDNs. iEDNs containing doxorubicin (iEDNs-DOX) showed a stronger inhibition of target tumor growth than untargeted ones. The resulting anti-EGFR iEDNs exhibited strong biocompatibility, prolonged blood circulation, and efficient targeting of TNBC in mice. Therefore, iEDNs may be used as potential TNBC-targeted co-delivery systems for therapeutics and diagnostics.
Collapse
|
34
|
Obozina AS, Komedchikova EN, Kolesnikova OA, Iureva AM, Kovalenko VL, Zavalko FA, Rozhnikova TV, Tereshina ED, Mochalova EN, Shipunova VO. Genetically Encoded Self-Assembling Protein Nanoparticles for the Targeted Delivery In Vitro and In Vivo. Pharmaceutics 2023; 15:231. [PMID: 36678860 PMCID: PMC9861179 DOI: 10.3390/pharmaceutics15010231] [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: 11/25/2022] [Revised: 12/30/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
Targeted nanoparticles of different origins are considered as new-generation diagnostic and therapeutic tools. However, there are no targeted drug formulations within the composition of nanoparticles approved by the FDA for use in the clinic, which is associated with the insufficient effectiveness of the developed candidates, the difficulties of their biotechnological production, and inadequate batch-to-batch reproducibility. Targeted protein self-assembling nanoparticles circumvent this problem since proteins are encoded in DNA and the final protein product is produced in only one possible way. We believe that the combination of the endless biomedical potential of protein carriers as nanoparticles and the standardized protein purification protocols will make significant progress in "magic bullet" creation possible, bringing modern biomedicine to a new level. In this review, we are focused on the currently existing platforms for targeted self-assembling protein nanoparticles based on transferrin, lactoferrin, casein, lumazine synthase, albumin, ferritin, and encapsulin proteins, as well as on proteins from magnetosomes and virus-like particles. The applications of these self-assembling proteins for targeted delivery in vitro and in vivo are thoroughly discussed, including bioimaging applications and different therapeutic approaches, such as chemotherapy, gene delivery, and photodynamic and photothermal therapy. A critical assessment of these protein platforms' efficacy in biomedicine is provided and possible problems associated with their further development are described.
Collapse
Affiliation(s)
| | | | | | - Anna M. Iureva
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Vera L. Kovalenko
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Fedor A. Zavalko
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | | | | | - Elizaveta N. Mochalova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
- Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Victoria O. Shipunova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
- Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia
| |
Collapse
|
35
|
Nikitin MP. Non-complementary strand commutation as a fundamental alternative for information processing by DNA and gene regulation. Nat Chem 2023; 15:70-82. [PMID: 36604607 DOI: 10.1038/s41557-022-01111-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 11/16/2022] [Indexed: 01/07/2023]
Abstract
The discovery of the DNA double helix has revolutionized our understanding of data processing in living systems, with the complementarity of the two DNA strands providing a reliable mechanism for the storage of hereditary information. Here I reveal the 'strand commutation' phenomenon-a fundamentally different mechanism of information storage and processing by DNA/RNA based on the reversible low-affinity interactions of essentially non-complementary nucleic acids. I demonstrate this mechanism by constructing a memory circuit, a 5-min square-root circuit for 4-bit inputs comprising only nine processing ssDNAs, simulating a 572-input AND gate (surpassing the bitness of current electronic computers), and elementary algebra systems with continuously changing variables. Most importantly, I show potential pathways of gene regulation with strands of maximum non-complementarity to the gene sequence that may be key to the reduction of off-target therapeutic effects. This Article uncovers the information-processing power of the low-affinity interactions that may underlie major processes in an organism-from short-term memory to cancer, ageing and evolution.
Collapse
Affiliation(s)
- Maxim P Nikitin
- Sirius University of Science and Technology, Sochi, Russia. .,Abisense LLC, Dolgoprudny, Moscow Region, Russia.
| |
Collapse
|
36
|
Kovalenko VL, Komedchikova EN, Sogomonyan AS, Tereshina ED, Kolesnikova OA, Mirkasymov AB, Iureva AM, Zvyagin AV, Nikitin PI, Shipunova VO. Lectin-Modified Magnetic Nano-PLGA for Photodynamic Therapy In Vivo. Pharmaceutics 2022; 15:pharmaceutics15010092. [PMID: 36678721 PMCID: PMC9862264 DOI: 10.3390/pharmaceutics15010092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/16/2022] [Accepted: 12/25/2022] [Indexed: 12/29/2022] Open
Abstract
The extreme aggressiveness and lethality of many cancer types appeal to the problem of the development of new-generation treatment strategies based on smart materials with a mechanism of action that differs from standard treatment approaches. The targeted delivery of nanoparticles to specific cancer cell receptors is believed to be such a strategy; however, there are no targeted nano-drugs that have successfully completed clinical trials to date. To meet the challenge, we designed an alternative way to eliminate tumors in vivo. Here, we show for the first time that the targeting of lectin-equipped polymer nanoparticles to the glycosylation profile of cancer cells, followed by photodynamic therapy (PDT), is a promising strategy for the treatment of aggressive tumors. We synthesized polymer nanoparticles loaded with magnetite and a PDT agent, IR775 dye (mPLGA/IR775). The magnetite incorporation into the PLGA particle structure allows for the quantitative tracking of their accumulation in different organs and the performing of magnetic-assisted delivery, while IR775 makes fluorescent in vivo bioimaging as well as light-induced PDT possible, thus realizing the theranostics concept. To equip PLGA nanoparticles with targeting modality, the particles were conjugated with lectins of different origins, and the flow cytometry screening revealed that the most effective candidate for breast cancer cell labeling is ConA, a lectin from Canavalia ensiformis. In vivo experiments showed that after i.v. administration, mPLGA/IR775-ConA nanoparticles efficiently accumulated in the allograft tumors under the external magnetic field; produced a bright fluorescent signal for in vivo bioimaging; and led to 100% tumor growth inhibition after the single session of PDT, even for large solid tumors of more than 200 mm3 in BALB/c mice. The obtained results indicate that the mPLGA/IR775 nanostructure has great potential to become a highly effective oncotheranostic agent.
Collapse
Affiliation(s)
- Vera L. Kovalenko
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701 Dolgoprudny, Russia
| | - Elena N. Komedchikova
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701 Dolgoprudny, Russia
| | - Anna S. Sogomonyan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia
| | - Ekaterina D. Tereshina
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701 Dolgoprudny, Russia
| | - Olga A. Kolesnikova
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701 Dolgoprudny, Russia
| | - Aziz B. Mirkasymov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia
| | - Anna M. Iureva
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701 Dolgoprudny, Russia
| | - Andrei V. Zvyagin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia
| | - Petr I. Nikitin
- Prokhorov General Physics Institute, Russian Academy of Sciences, 38 Vavilov Street, 119991 Moscow, Russia
| | - Victoria O. Shipunova
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701 Dolgoprudny, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia
- Nanobiomedicine Division, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia
- Correspondence:
| |
Collapse
|
37
|
Komedchikova EN, Kolesnikova OA, Tereshina ED, Kotelnikova PA, Sogomonyan AS, Stepanov AV, Deyev SM, Nikitin MP, Shipunova VO. Two-Step Targeted Drug Delivery via Proteinaceous Barnase-Barstar Interface and Doxorubicin-Loaded Nano-PLGA Outperforms One-Step Strategy for Targeted Delivery to HER2-Overexpressing Cells. Pharmaceutics 2022; 15:52. [PMID: 36678681 PMCID: PMC9861000 DOI: 10.3390/pharmaceutics15010052] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Nanoparticle-based chemotherapy is considered to be an effective approach to cancer diagnostics and therapy in modern biomedicine. However, efficient tumor targeting remains a great challenge due to the lack of specificity, selectivity, and high dosage of chemotherapeutic drugs required. A two-step targeted drug delivery strategy (DDS), involving cancer cell pre-targeting, first with a first nontoxic module and subsequent targeting with a second complementary toxic module, is a solution for decreasing doses for administration and lowering systemic toxicity. To prove two-step DDS efficiency, we performed a direct comparison of one-step and two-step DDS based on chemotherapy loaded PLGA nanoparticles and barnase*barstar interface. Namely, we developed and thoroughly characterized the two-step targeting strategy of HER2-overexpressing cancer cells. The first targeting block consists of anti-HER2 scaffold polypeptide DARPin9_29 fused with barstar. Barstar exhibits an extremely effective binding to ribonuclease barnase with Kaff = 1014 M-1, thus making the barnase*barstar protein pair one of the strongest known protein*protein complexes. A therapeutic PLGA-based nanocarrier coupled to barnase was used as a second targeting block. The PLGA nanoparticles were loaded with diagnostic dye, Nile Blue, and a chemotherapeutic drug, doxorubicin. We showed that the two-step DDS increases the performance of chemotherapy-loaded nanocarriers: IC50 of doxorubicin delivered via two-step DDS was more than 100 times lower than that for one-step DDS: IC50 = 43 ± 3 nM for two-step DDS vs. IC50 = 4972 ± 1965 nM for one-step DDS. The obtained results demonstrate the significant efficiency of two-step DDS over the classical one-step one. We believe that the obtained data will significantly change the direction of research in developing targeted anti-cancer drugs and promote the creation of new generation cancer treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Polina A. Kotelnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Anna S. Sogomonyan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Alexey V. Stepanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Sergey M. Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Maxim P. Nikitin
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Victoria O. Shipunova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia
| |
Collapse
|
38
|
Drozdov AS, Komarova KS, Mochalova EN, Komedchikova EN, Shipunova VO, Nikitin MP. Fluorescent Magnetic Nanoparticles for Bioimaging through Biomimetic Surface Modification. Int J Mol Sci 2022; 24:ijms24010134. [PMID: 36613578 PMCID: PMC9820170 DOI: 10.3390/ijms24010134] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Nanostructured materials and systems find various applications in biomedical fields. Hybrid organo-inorganic nanomaterials are intensively studied in a wide range of areas, from visualization to drug delivery or tissue engineering. One of the recent trends in material science is biomimetic approaches toward the synthesis or modification of functional nanosystems. Here, we describe an approach toward multifunctional nanomaterials through the biomimetic polymerization of dopamine derivatives. Magnetite nanoparticles were modified with a combination of dopamine conjugates to give multifunctional magneto-fluorescent nanocomposites in one synthetic step. The obtained material showed excellent biocompatibility at concentrations up to 200 μg/mL and an in vivo biodistribution profile typical for nanosized formulations. The synthesized systems were conjugated with antibodies against HER2 to improve their selectivity toward HER2-positive cancer cells. The produced material can be used for dual magneto-optical in vivo studies or targeted drug delivery. The applied synthetic strategy can be used for the creation of various multifunctional hybrid nanomaterials in mild conditions.
Collapse
Affiliation(s)
- Andrey S Drozdov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Moscow Region, Russia
| | - Kristina S Komarova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Moscow Region, Russia
| | - Elizaveta N Mochalova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Moscow Region, Russia
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991 Moscow, Russia
- Research Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Elena N Komedchikova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Moscow Region, Russia
| | - Victoria O Shipunova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Moscow Region, Russia
- Research Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Maxim P Nikitin
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Moscow Region, Russia
- Research Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
39
|
Kotelnikova PA, Iureva AM, Nikitin MP, Zvyagin AV, Deyev SM, Shipunova VO. Peroxidase-like activity of silver nanowires and its application for colorimetric detection of the antibiotic chloramphenicol. TALANTA OPEN 2022. [DOI: 10.1016/j.talo.2022.100164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
40
|
Zelepukin IV, Griaznova OY, Shevchenko KG, Ivanov AV, Baidyuk EV, Serejnikova NB, Volovetskiy AB, Deyev SM, Zvyagin AV. Flash drug release from nanoparticles accumulated in the targeted blood vessels facilitates the tumour treatment. Nat Commun 2022; 13:6910. [PMID: 36376302 PMCID: PMC9661469 DOI: 10.1038/s41467-022-34718-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022] Open
Abstract
Tumour microenvironment hinders nanoparticle transport deep into the tissue precluding thorough treatment of solid tumours and metastatic nodes. We introduce an anticancer drug delivery concept termed FlaRE (Flash Release in Endothelium), which represents alternative to the existing approaches based on enhanced permeability and retention effect. This approach relies on enhanced drug-loaded nanocarrier accumulation in vessels of the target tumour or metastasised organ, followed by a rapid release of encapsulated drug within tens of minutes. It leads to a gradient-driven permeation of the drug to the target tissue. This pharmaceutical delivery approach is predicted by theoretical modelling and validated experimentally using rationally designed MIL-101(Fe) metal-organic frameworks. Doxorubicin-loaded MIL-101 nanoparticles get swiftly trapped in the vasculature of the metastasised lungs, disassemble in the blood vessels within 15 minutes and release drug, which rapidly impregnates the organ. A significant improvement of the therapeutic outcome is demonstrated in animal models of early and late-stage B16-F1 melanoma metastases with 11-fold and 4.3-fold decrease of pulmonary melanoma nodes, respectively.
Collapse
Affiliation(s)
- Ivan V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia.
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409, Moscow, Russia.
| | - Olga Yu Griaznova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409, Moscow, Russia
| | - Konstantin G Shevchenko
- Institute of Cytology of the Russian Academy of Sciences, 194064, Saint Petersburg, Russia
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Drugs of the Russian Academy of Sciences, 108819, Moscow, Russia
| | - Andrey V Ivanov
- Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Ekaterina V Baidyuk
- Institute of Cytology of the Russian Academy of Sciences, 194064, Saint Petersburg, Russia
| | - Natalia B Serejnikova
- Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Artur B Volovetskiy
- Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia.
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409, Moscow, Russia.
- Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia.
| | - Andrei V Zvyagin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia.
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409, Moscow, Russia.
- Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia.
- MQ Photonics Centre, Macquarie University, 2109, Sydney, Australia.
| |
Collapse
|
41
|
Egorova EA, Nikitin MP. Delivery of Theranostic Nanoparticles to Various Cancers by Means of Integrin-Binding Peptides. Int J Mol Sci 2022; 23:ijms232213735. [PMID: 36430214 PMCID: PMC9696485 DOI: 10.3390/ijms232213735] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Abstract
Active targeting of tumors is believed to be the key to efficient cancer therapy and accurate, early-stage diagnostics. Active targeting implies minimized off-targeting and associated cytotoxicity towards healthy tissue. One way to acquire active targeting is to employ conjugates of therapeutic agents with ligands known to bind receptors overexpressed onto cancer cells. The integrin receptor family has been studied as a target for cancer treatment for almost fifty years. However, systematic knowledge on their effects on cancer cells, is yet lacking, especially when utilized as an active targeting ligand for particulate formulations. Decoration with various integrin-targeting peptides has been reported to increase nanoparticle accumulation in tumors ≥ 3-fold when compared to passively targeted delivery. In recent years, many newly discovered or rationally designed integrin-binding peptides with excellent specificity towards a single integrin receptor have emerged. Here, we show a comprehensive analysis of previously unreviewed integrin-binding peptides, provide diverse modification routes for nanoparticle conjugation, and showcase the most notable examples of their use for tumor and metastases visualization and eradication to date, as well as possibilities for combined cancer therapies for a synergetic effect. This review aims to highlight the latest advancements in integrin-binding peptide development and is directed to aid transition to the development of novel nanoparticle-based theranostic agents for cancer therapy.
Collapse
Affiliation(s)
- Elena A. Egorova
- Department of Nanobiomedicine, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sirius, Russia
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 1 Meditsinskaya Str., 603081 Nizhny Novgorod, Russia
| | - Maxim P. Nikitin
- Department of Nanobiomedicine, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sirius, Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy per., 141701 Dolgoprudny, Russia
- Correspondence:
| |
Collapse
|
42
|
Stimuli-responsive nanoassemblies for targeted delivery against tumor and its microenvironment. Biochim Biophys Acta Rev Cancer 2022; 1877:188779. [PMID: 35977690 DOI: 10.1016/j.bbcan.2022.188779] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 02/06/2023]
Abstract
Despite the emergence of various cancer treatments, such as surgery, chemotherapy, radiotherapy, and immunotherapy, their use remains restricted owing to their limited tumor elimination efficacy and side effects. The use of nanoassemblies as delivery systems in nanomedicine for tumor diagnosis and therapy is flourishing. These nanoassemblies can be designed to have various shapes, sizes, and surface charges to meet the requirements of different applications. It is crucial for nanoassemblies to have enhanced delivery of payloads while inducing minimal to no toxicity to healthy tissues. In this review, stimuli-responsive nanoassemblies capable of combating the tumor microenvironment (TME) are discussed. First, various TME characteristics, such as hypoxia, oxidoreduction, adenosine triphosphate (ATP) elevation, and acidic TME, are described. Subsequently, the unique characteristics of the vascular and stromal TME are differentiated, and multiple barriers that have to be overcome are discussed. Furthermore, strategies to overcome these barriers for successful drug delivery to the targeted site are reviewed and summarized. In conclusion, the possible challenges and prospects of using these nanoassemblies for tumor-targeted delivery are discussed. This review aims at inspiring researchers to develop stimuli-responsive nanoassemblies for tumor-targeted delivery for clinical applications.
Collapse
|
43
|
Shivanna AT, Dash BS, Chen JP. Functionalized Magnetic Nanoparticles for Alternating Magnetic Field- or Near Infrared Light-Induced Cancer Therapies. MICROMACHINES 2022; 13:1279. [PMID: 36014201 PMCID: PMC9413965 DOI: 10.3390/mi13081279] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/02/2022] [Accepted: 08/06/2022] [Indexed: 05/14/2023]
Abstract
The multi-faceted nature of functionalized magnetic nanoparticles (fMNPs) is well-suited for cancer therapy. These nanocomposites can also provide a multimodal platform for targeted cancer therapy due to their unique magnetic guidance characteristics. When induced by an alternating magnetic field (AMF), fMNPs can convert the magnetostatic energy to heat for magnetic hyperthermia (MHT), as well as for controlled drug release. Furthermore, with the ability to convert near-infrared (NIR) light energy to heat energy, fMNPs have attracted interest for photothermal therapy (PTT). Other than MHT and PTT, fMNPs also have a place in combination cancer therapies, such as chemo-MHT, chemo-PTT, and chemo-PTT-photodynamic therapy, among others, due to their versatile properties. Thus, this review presents multifunctional nanocomposites based on fMNPs for cancer therapies, induced by an AMF or NIR light. We will first discuss the different fMNPs induced with an AMF for cancer MHT and chemo-MHT. Secondly, we will discuss fMNPs irradiated with NIR lasers for cancer PTT and chemo-PTT. Finally, fMNPs used for dual-mode AMF + NIR-laser-induced magneto-photo-hyperthermia (MPHT) will be discussed.
Collapse
Affiliation(s)
| | - Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33305, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
44
|
Tao Y, Lan X, Zhang Y, Xiao Y, Wang J, Chen H, Liu L, Liang XJ, Guo W. Navigations of the targeting pathway of nanomedicines towards tumor. Expert Opin Drug Deliv 2022; 19:985-996. [PMID: 35929954 DOI: 10.1080/17425247.2022.2110064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Nanomedicines (NMs) have emerged as a promising approach for revolutionizing cancer treatment outcomes, mainly due to their benefits in the tumor targeted delivery of therapeutics. The preferential accumulation of NMs in tumor has been widely verified by macroscopical technologies. Accordingly, several classic and emerging targeting mechanisms have been proposed to support the tumor-specific delivery of NMs. The targeting mechanism has been a topic of intensive interest and controversy in the field of NMs development. Especially, the mechanisms by which NMs target tumor remain elusive. AREA COVERED This topical review mainly discussed the evolution of the targeting mechanisms, crucial issues associated with each mechanism, and confused debates among the mechanisms. The targeting mechanisms of tumor-specific NMs discussed here include the enhanced permeability and retention (EPR) effect, protein corona-mediated targeting delivery, circulating cell mediated transportation, and transcytosis. EXPERT OPINION It is of great significance for ultimate clinical translation to have more comprehensive considerations on the mechanism driving the pathway of NMs toward tumors. Our thoughts in this review are expected to provide comprehensive understanding on the mechanisms and elicit thorough explorations on new mechanism to renovate the knowledge framework of NMs delivery.
Collapse
Affiliation(s)
- Ying Tao
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, College of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, P. R. China
| | - Xinmiao Lan
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Yuxuan Zhang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yafang Xiao
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, College of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, P. R. China
| | - Jinjin Wang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Haoting Chen
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, College of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, P. R. China
| | - Lu Liu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Xing-Jie Liang
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, College of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, P. R. China.,Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, College of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, P. R. China
| |
Collapse
|
45
|
Nanocarriers: A Reliable Tool for the Delivery of Anticancer Drugs. Pharmaceutics 2022; 14:pharmaceutics14081566. [PMID: 36015192 PMCID: PMC9415391 DOI: 10.3390/pharmaceutics14081566] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/26/2022] Open
Abstract
Nanomedicines have gained popularity due to their potential therapeutic applications, especially cancer treatment. Targeted nanoparticles can deliver drugs directly to cancer cells and enable prolonged drug release, reducing off-target toxicity and increasing therapeutic efficacy. However, translating nanomedicines from preclinical to clinical settings has been difficult. Rapid advancements in nanotechnology promise to enhance cancer therapies. Nanomedicine offers advanced targeting and multifunctionality. Nanoparticles (NPs) have several uses nowadays. They have been studied as drug transporters, tumor gene delivery agents, and imaging contrast agents. Nanomaterials based on organic, inorganic, lipid, or glycan substances and synthetic polymers have been used to enhance cancer therapies. This review focuses on polymeric nanoparticle delivery strategies for anticancer nanomedicines.
Collapse
|
46
|
Song X, Fu W, Cheang UK. Immunomodulation and delivery of macrophages using nano-smooth drug-loaded magnetic microrobots for dual targeting cancer therapy. iScience 2022; 25:104507. [PMID: 35720266 PMCID: PMC9201018 DOI: 10.1016/j.isci.2022.104507] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 04/04/2022] [Accepted: 05/27/2022] [Indexed: 11/24/2022] Open
Abstract
To realize the potential to use micro/nanorobots for targeted cancer therapy, it is important to improve their biocompatibility and targeting ability. Here, we report on drug-loaded magnetic microrobots capable of polarizing macrophages into the antitumor phenotype to target and inhibit cancer cells. In vitro tests demonstrated that the microrobots have good biocompatibility with normal cells and immune cells. Positively charged DOX was loaded onto the surface of microrobots via electrostatic interactions and exhibited pH-responsive release behavior. The nano-smooth surfaces of the microrobots activated M1 polarization of macrophages, thus activating their intrinsic targeting and antitumor abilities toward cancer cells. Through dual targeting from magnetic guidance and M1 macrophages, the microrobots were able to target and kill cancer cells in a 3D tumor spheroid culture assay. These findings demonstrate a way to improve the tumor-targeting and antitumor abilities of microrobots through the combined use of magnetic control, macrophages, and pH-responsive drug release.
Collapse
Affiliation(s)
- Xiaoxia Song
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wei Fu
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - U Kei Cheang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen 518055, China
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Southern University of Science and Technology, Shenzhen, China
- Guangdong Provincial Key Laboratory of Human-Augmentation and Rehabilitation Robotics in Universities, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
47
|
Red Blood Cell Inspired Strategies for Drug Delivery: Emerging Concepts and New Advances. Pharm Res 2022; 39:2673-2698. [PMID: 35794397 DOI: 10.1007/s11095-022-03328-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/29/2022] [Indexed: 12/09/2022]
Abstract
In the past five decades, red blood cells (RBCs) have been extensively explored as drug delivery systems due to their distinguishing potential in modulating the pharmacokinetic, pharmacodynamics, and biological activity of carried payloads. The extensive interests in RBC-mediated drug delivery technologies are in part derived from RBCs' unique biological features such as long circulation time, wide access to many tissues in the body, and low immunogenicity. Owing to these outstanding properties, a large body of efforts have led to the development of various RBC-inspired strategies to enable precise drug delivery with enhanced therapeutic efficacy and reduced off-target toxicity. In this review, we discuss emerging concepts and new advances in such RBC-inspired strategies, including native RBCs, ghost RBCs, RBC-mimetic nanoparticles, and RBC-derived extracellular vesicles, for drug delivery.
Collapse
|
48
|
Lu Z, Singh G, Lesani P, Zreiqat H. Promise and Perspective of Nanomaterials in Antisenescence Tissue Engineering Applications. ACS Biomater Sci Eng 2022; 8:3133-3141. [PMID: 35771746 DOI: 10.1021/acsbiomaterials.1c01298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The tissue engineering approach for repair and regeneration has achieved significant progress over the past decades. However, challenges remain in developing strategies to solve the declined or impaired innate cell and tissue regeneration capacity that occurs with aging. Cellular senescence is a key mechanism underlying organismal aging and is responsible for the declined tissue regeneration capacity in the aging population. Therefore, to promote the diminished tissue regeneration ability in the aged population, it is critical to developing a feasible and promising strategy to target senescent cells. Recent advances in nanomaterials have revolutionized biomedical applications ranging from biosensing to bioimaging and targeted drug delivery. In this perspective, we review and discuss the nature and influences of cell-intrinsic and cell-extrinsic factors on reduced regenerative abilities through aging and how nanotechnology can be a therapeutic avenue to sense, rejuvenate, and eliminate senescent cells, thereby improving the tissue regeneration capacity in the aging population.
Collapse
Affiliation(s)
- ZuFu Lu
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,ARC Training Centre for Innovative BioEngineering, The University of Sydney, Sydney New South Wales 2006, Australia
| | - Gurvinder Singh
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,ARC Training Centre for Innovative BioEngineering, The University of Sydney, Sydney New South Wales 2006, Australia
| | - Pooria Lesani
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,ARC Training Centre for Innovative BioEngineering, The University of Sydney, Sydney New South Wales 2006, Australia
| | - Hala Zreiqat
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,ARC Training Centre for Innovative BioEngineering, The University of Sydney, Sydney New South Wales 2006, Australia
| |
Collapse
|
49
|
Nowak-Jary J, Machnicka B. Pharmacokinetics of magnetic iron oxide nanoparticles for medical applications. J Nanobiotechnology 2022; 20:305. [PMID: 35761279 PMCID: PMC9235206 DOI: 10.1186/s12951-022-01510-w] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/07/2022] [Indexed: 12/05/2022] Open
Abstract
Magnetic iron oxide nanoparticles (MNPs) have been under intense investigation for at least the last five decades as they show enormous potential for many biomedical applications, such as biomolecule separation, MRI imaging and hyperthermia. Moreover, a large area of research on these nanostructures is concerned with their use as carriers of drugs, nucleic acids, peptides and other biologically active compounds, often leading to the development of targeted therapies. The uniqueness of MNPs is due to their nanometric size and unique magnetic properties. In addition, iron ions, which, along with oxygen, are a part of the MNPs, belong to the trace elements in the body. Therefore, after digesting MNPs in lysosomes, iron ions are incorporated into the natural circulation of this element in the body, which reduces the risk of excessive storage of nanoparticles. Still, one of the key issues for the therapeutic applications of magnetic nanoparticles is their pharmacokinetics which is reflected in the circulation time of MNPs in the bloodstream. These characteristics depend on many factors, such as the size and charge of MNPs, the nature of the polymers and any molecules attached to their surface, and other. Since the pharmacokinetics depends on the resultant of the physicochemical properties of nanoparticles, research should be carried out individually for all the nanostructures designed. Almost every year there are new reports on the results of studies on the pharmacokinetics of specific magnetic nanoparticles, thus it is very important to follow the achievements on this matter. This paper reviews the latest findings in this field. The mechanism of action of the mononuclear phagocytic system and the half-lives of a wide range of nanostructures are presented. Moreover, factors affecting clearance such as hydrodynamic and core size, core morphology and coatings molecules, surface charge and technical aspects have been described.
Collapse
Affiliation(s)
- Julia Nowak-Jary
- Department of Biotechnology, Institute of Biological Sciences, University of Zielona Gora, Prof. Z. Szafrana 1, 65-516, Zielona Gora, Poland.
| | - Beata Machnicka
- Department of Biotechnology, Institute of Biological Sciences, University of Zielona Gora, Prof. Z. Szafrana 1, 65-516, Zielona Gora, Poland
| |
Collapse
|
50
|
Garello F, Svenskaya Y, Parakhonskiy B, Filippi M. Micro/Nanosystems for Magnetic Targeted Delivery of Bioagents. Pharmaceutics 2022; 14:pharmaceutics14061132. [PMID: 35745705 PMCID: PMC9230665 DOI: 10.3390/pharmaceutics14061132] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/09/2022] [Accepted: 05/19/2022] [Indexed: 01/09/2023] Open
Abstract
Targeted delivery of pharmaceuticals is promising for efficient disease treatment and reduction in adverse effects. Nano or microstructured magnetic materials with strong magnetic momentum can be noninvasively controlled via magnetic forces within living beings. These magnetic carriers open perspectives in controlling the delivery of different types of bioagents in humans, including small molecules, nucleic acids, and cells. In the present review, we describe different types of magnetic carriers that can serve as drug delivery platforms, and we show different ways to apply them to magnetic targeted delivery of bioagents. We discuss the magnetic guidance of nano/microsystems or labeled cells upon injection into the systemic circulation or in the tissue; we then highlight emergent applications in tissue engineering, and finally, we show how magnetic targeting can integrate with imaging technologies that serve to assist drug delivery.
Collapse
Affiliation(s)
- Francesca Garello
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy;
| | - Yulia Svenskaya
- Science Medical Center, Saratov State University, 410012 Saratov, Russia;
| | - Bogdan Parakhonskiy
- Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium;
| | - Miriam Filippi
- Soft Robotics Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland
- Correspondence:
| |
Collapse
|