1
|
Abdullah KM, Sharma G, Singh AP, Siddiqui JA. Nanomedicine in Cancer Therapeutics: Current Perspectives from Bench to Bedside. Mol Cancer 2025; 24:169. [PMID: 40490771 DOI: 10.1186/s12943-025-02368-w] [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: 03/26/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
Cancer is among the leading causes of death worldwide, with projections indicating that it will claim 35 million lives by the year 2050. Conventional therapies, such as chemotherapy and immune modulation, have reduced cancer mortality to some extent; however, they have limited efficacy due to their broad mode of action, often resulting in cytotoxic effects on normal cells along with the malignant tissues, ultimately limiting their overall optimal therapeutic efficacy outcomes.Rapid advances in nanotechnology and an evolving understanding of cancer mechanisms have propelled the development of a diverse array of nanocarriers to vanquish the hurdles in achieving sophisticated drug delivery with reduced off-target toxicity. Nanoformulations can deliver the anti-cancer agents precisely to the tumor cell by integrating a multitarget approach that allows for tissue-, cell-, or organelle-specific delivery and internalization. Despite the immense interest and unmatched advancements in modern oncology equipped with nanomedicines, only a few nanoformulations have successfully translated into clinical settings. A major reason behind this shortcoming is the lack of a rationale design incorporating smart, responsive targeting features, leading to a compromised therapeutic window due to inefficient internalization or erroneous intracellular localization with unsuccessful payload release. This review aims to summarize the recent perspective of nanomedicine and its translation to clinical practice, with a particular focus on the evolution of strategies used in tumor targeting from traditional EPR-based passive mechanisms to advanced active and multi-stage approaches. We highlight the coupling of organelle-specific and stimuli-responsive nanocarriers, discuss the potential of biomimetic and cell-mediated delivery systems, and also shed light on technologies such as microfluidics, tumor-on-chip models, and AI-assisted synthesis. Finally, this review explores translational hurdles ranging from biological and manufacturing challenges to regulatory bottlenecks and outlines how innovative modeling systems and engineering solutions can bridge the gap from bench to bedside in cancer nanotherapeutics.
Collapse
Affiliation(s)
- K M Abdullah
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Gunjan Sharma
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Ajay P Singh
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Jawed A Siddiqui
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
| |
Collapse
|
2
|
Mostafavi M, Ghazi F, Mehrabifard M, Alivirdiloo V, Hajiabbasi M, Rahimi F, Mobed A, Taheripak G, Ramezani Farani M, Huh YS, Bakhtiyari S, Alipourfard I. State-of-the-art application of nanoparticles in radiotherapy: a platform for synergistic effects in cancer treatment. Strahlenther Onkol 2025; 201:577-588. [PMID: 39367110 PMCID: PMC12119707 DOI: 10.1007/s00066-024-02301-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/24/2024] [Indexed: 10/06/2024]
Abstract
Radiotherapy (RT) is a gold standard cancer treatment worldwide. However, RT has limitations and many side effects. Nanoparticles (NPs) have exclusive properties that allow them to be used in cancer therapy. Consequently, the combination of NP and RT opens up a new frontier in cancer treatment. Among NPs, gold nanoparticles (GNPs) are the most extensively studied and are considered ideal radiosensitizers for radiotherapy due to their unique physicochemical properties and high X‑ray absorption. This review analyzes the various roles of NPs as radiosensitizers in radiotherapy of glioblastoma (GBS), prostate cancer, and breast cancer and summarizes recent advances. Furthermore, the underlying mechanisms of NP radiosensitization, including physical, chemical, and biological mechanisms, are discussed, which may provide new directions for next-generation GNP optimization and clinical transformation.
Collapse
Affiliation(s)
- Mehrnaz Mostafavi
- Faculty of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farhood Ghazi
- Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Vahid Alivirdiloo
- Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | | | - Fatemeh Rahimi
- Division of Clinical Laboratory, Zahra Mardani Azar Children Training Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mobed
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gholamreza Taheripak
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon, Korea (Republic of)
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon, Korea (Republic of)
| | - Salar Bakhtiyari
- Department of Clinical Biochemistry, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Iraj Alipourfard
- Iraj Alipourfard, Institute of Physical Chemistry, Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224, Warsaw, Poland.
| |
Collapse
|
3
|
Belyaev IB, Griaznova OY, Yaremenko AV, Deyev SM, Zelepukin IV. Beyond the EPR effect: Intravital microscopy analysis of nanoparticle drug delivery to tumors. Adv Drug Deliv Rev 2025; 219:115550. [PMID: 40021012 DOI: 10.1016/j.addr.2025.115550] [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: 10/31/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Delivery of nanoparticles (NPs) to solid tumors has long relied on enhanced permeability and retention (EPR) effect, involving permeation of NPs through a leaky vasculature with prolonged retention by reduced lymphatic drainage in tumor. Recent research studies and clinical data challenge EPR concept, revealing alternative pathways and approaches of NP delivery. The area was significantly impacted by the implementation of intravital optical microscopy, unraveling delivery mechanisms at cellular level in vivo. This review presents analysis of the reasons for EPR heterogeneity in tumors and describes non-EPR based concepts for drug delivery, which can supplement the current paradigm. One of the approaches is targeting tumor endothelium by NPs with subsequent intravascular drug release and gradient-driven drug transport to tumor interstitium. Others exploit various immune cells for tumor infiltration and breaking endothelial barriers. Finally, we discuss the involvement of active transcytosis through endothelial cells in NP delivery. This review aims to inspire further understanding of the process of NP extravasation in tumors and provide insights for developing next-generation nanomedicines with improved delivery.
Collapse
Affiliation(s)
- Iaroslav B Belyaev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Olga Yu Griaznova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Ivan V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75123, Sweden.
| |
Collapse
|
4
|
Tang J, Zhang J, Li Y, Hu Y, He D, Ni H, Zhang J, Wu F, Tang Y, Wang S. Interpretable Radiomics Model Predicts Nanomedicine Tumor Accumulation Using Routine Medical Imaging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2416696. [PMID: 39916575 DOI: 10.1002/adma.202416696] [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: 10/30/2024] [Revised: 12/18/2024] [Indexed: 03/27/2025]
Abstract
Accurately predicting nanomedicine accumulation is critical for guiding patient stratification and optimizing treatment strategies in the context of precision medicine. However, non-invasive prediction of nanomedicine accumulation remains challenging, primarily due to the complexity of identifying relevant imaging features that predict accumulation. Here, a novel non-invasive method is proposed that utilizes standard-of-care medical imaging modalities, including computed tomography and ultrasound, combined with a radiomics-based model to predict nanomedicine accumulation in tumor. The model is validated using a test dataset consisting of seven tumor xenografts in mice and three sizes of gold nanoparticles, achieving an area under the receiver operating characteristic curve of 0.851. The median accumulation levels of tumors predicted as "high accumulators" are 2.69 times greater than those predicted as "low accumulators". Analysis of this machine-learning-driven interpretable radiomics model revealed imaging features that are strongly correlated with dense stroma, a recognized biological barrier to effective nanomedicine delivery. Radiomics-based prediction of tumor accumulation holds promise for stratifying patient and enabling precise tailoring of nanomedicine treatment strategies.
Collapse
Affiliation(s)
- Jiajia Tang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Jie Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Yang Li
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Yongzhi Hu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Doudou He
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Hao Ni
- Department of Mathematics, University College London, London, WC1H0AY, UK
| | - Jiulou Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Feiyun Wu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Yuxia Tang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Shouju Wang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| |
Collapse
|
5
|
Gupta S, Kaur R, Bhardwaj A, Parashar D. Multifunctional Nanomaterials: Recent Advancements in Cancer Therapeutics and Vaccines. Indian J Microbiol 2025; 65:51-68. [PMID: 40371018 PMCID: PMC12069785 DOI: 10.1007/s12088-024-01274-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/27/2024] [Indexed: 05/16/2025] Open
Abstract
Nanotechnology has revolutionized cancer detection and treatment, overcoming limitations of conventional methods. Imaging, targeting, and therapy moieties can all be combined in multifunctional nanoparticle systems to deliver the imaging or treatment modalities to the tumor in a targeted manner. These nanostructures can be engineered to create smart drug delivery systems for effective distribution and combinatorial therapy. Nanostructures made of biomolecules are naturally multifunctional and have a variety of biological functions that can be investigated for use in cancer nanomedicine. The supramolecular characteristics of biomolecules can be carefully engineered to create smart drug delivery systems that enable effective drug distribution to specific areas of the body as well as combinatorial therapy in a single design. Nanotechnology has also increased the efficiency of cancer vaccines, highlighting the future of tumor immunotherapy. Nanomaterials are often used as anti-cancer drugs or anti-inflammatory drugs due to their biosafety potential and enhanced bioavailability. By delivering targeted antigens and adjuvants, nanomaterials can improve vaccination efficacy and safety, preventing rapid degradation and prolonging antigen retention in lymphoid and tumor cells. We examine both organic and inorganic multifunctional nanomaterials in this review, emphasizing particular multifunctional properties in the context of cancer targeting, therapy, and vaccinations.
Collapse
Affiliation(s)
- Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh 281406 India
| | - Rasanpreet Kaur
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh 281406 India
| | - Alok Bhardwaj
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh 281406 India
| | - Deepak Parashar
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226 United States
| |
Collapse
|
6
|
El-Sahli S, Manturthi S, Durocher E, Bo Y, Akman A, Sannan C, Kirkby M, Iroakazi CD, Deyell H, Kaczmarek S, Lee SH, Iqbal U, Côté M, Wang L, Gadde S. Nanoparticle-Mediated mRNA Delivery to Triple-Negative Breast Cancer (TNBC) Patient-Derived Xenograft (PDX) Tumors. ACS Pharmacol Transl Sci 2025; 8:460-469. [PMID: 39974646 PMCID: PMC11833720 DOI: 10.1021/acsptsci.4c00597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 02/21/2025]
Abstract
mRNA-based therapies can overcome several challenges faced by traditional therapies in treating a variety of diseases by selectively modulating genes and proteins without genomic integration. However, due to mRNA's poor stability and inherent limitations, nanoparticle (NP) platforms have been developed to deliver functional mRNA into cells. In cancer treatment, mRNA technology has multiple applications, such as restoration of tumor suppressors and activating antitumor immunity. Most of these applications have been evaluated using simple cell-line-based tumor models, which failed to represent the complexity, heterogeneity, and 3D architecture of patient tumors. This discrepancy has led to inconsistencies and failures in clinical translation. Compared to cell line models, patient-derived xenograft (PDX) models more accurately represent patient tumors and are better suitable for modeling. Therefore, for the first time, this study employed two different TNBC PDX tumors to examine the effects of the mRNA-NPs. mRNA-NPs are developed using EGFP-mRNA as a model and studied in TNBC cell lines, ex vivo TNBC PDX organotypic slice cultures, and in vivo TNBC PDX tumors. Our findings show that NPs can effectively accumulate in tumors after intravenous administration, protecting and delivering mRNA to PDX tumors with different genetic and chemosensitivity backgrounds. These studies offer more clinically relevant modeling systems for mRNA nanotherapies in cancer applications.
Collapse
Affiliation(s)
- Sara El-Sahli
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Shireesha Manturthi
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, ON K1Y 1J8, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Emma Durocher
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, ON K1Y 1J8, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Yuxia Bo
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, ON K1Y 1J8, Canada
| | - Alexandra Akman
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Christina Sannan
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Melanie Kirkby
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Chiamaka Divine Iroakazi
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Hannah Deyell
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Shelby Kaczmarek
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Seung-Hwan Lee
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Umar Iqbal
- Human
Health
Therapeutics Research Centre, National Research Council Canada, Ottawa K1A 0R6, Canada
| | - Marceline Côté
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Lisheng Wang
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Suresh Gadde
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, ON K1Y 1J8, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa-Carleton
Institute for Biomedical Engineering (OCIBME), Ottawa K1S 5B6, Canada
| |
Collapse
|
7
|
Chakraborty DD, Chakraborty P, Mondal A. An insight into cancer nanomedicine based on polysaccharides. Int J Biol Macromol 2025; 290:138678. [PMID: 39672407 DOI: 10.1016/j.ijbiomac.2024.138678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/29/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
With cancer rates on the rise around the world, cancer treatment has dominated scientific discussions in recent years. The toxicity of cytotoxic drugs, their lack of tumor localization, and their uniform dispersion into tumor tissues are the obstacles to cancer therapy. Other cancer treatment drawbacks include short blood circulation half-lives and undesirable pharmacokinetic behavior. Low-molecular-weight drugs conjugated with macromolecular carriers are better distributed in the body. The enhanced permeation and retention (EPR) effect causes natural and synthetic polymers, such as polysaccharides, proteins, antibodies, and poly amino acids, to accumulate in tumor tissue. Many manufactured and natural polymers are attractive polymeric drug carriers, allowing the creation of prodrugs from medicinal substances. Polysaccharides are biological polymers with structural and functional variations. They are also non-toxic, hydrophilic, biodegradable, and efficiently bioactive. Polysaccharides are ideal for synthesizing many nanoparticles due to their functional groups. Their ability to adapt to their microenvironment makes them valuable. Nanoplatforms based on polysaccharides can deliver targeted anticancer drugs for personalized cancer treatment. Unique polysaccharide structures and properties offer chemical and biological advantages for novel drug delivery. Polysaccharide-drug conjugation could revolutionize cancer chemotherapy. This study investigates polysaccharide conjugates and polysaccharides as natural biomaterials for cancer drug delivery.
Collapse
Affiliation(s)
| | - Prithviraj Chakraborty
- Royal School of Pharmacy, The Assam Royal Global University, Betkuchi, Guwahati-781035, India
| | - Arijit Mondal
- Department of Pharmaceutical Chemistry, M.R. College of Pharmaceutical Sciences and Research, Balisha-743234, India.
| |
Collapse
|
8
|
Li R, Huang J, Wei Y, Wang Y, Lu C, Liu J, Ma X. Nanotherapeutics for Macrophage Network Modulation in Tumor Microenvironments: Targets and Tools. Int J Nanomedicine 2024; 19:13615-13651. [PMID: 39717515 PMCID: PMC11665441 DOI: 10.2147/ijn.s491573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/04/2024] [Indexed: 12/25/2024] Open
Abstract
Macrophage is an important component in the tumor immune microenvironment, which exerts significant influence on tumor development and metastasis. Due to their dual nature of promoting and suppressing inflammation, macrophages can serve as both targets for tumor immunotherapy and tools for treating malignancies. However, the abundant infiltration of tumor-associated macrophages dominated by an immunosuppressive phenotype maintains a pro-tumor microenvironment, and engineering macrophages using nanotechnology to manipulate the tumor immune microenvironment represent a feasible approach for cancer immunotherapy. Additionally, considering the phagocytic and specifically tumor-targeting capabilities of M1 macrophages, macrophages manipulated through cellular engineering and nanotechnology, as well as macrophage-derived exosomes and macrophage membranes, can also become effective tools for cancer treatment. In conclusion, nanotherapeutics targeting macrophages remains immense potential for the development of macrophage-mediated tumor treatment methods and will further enhance our understanding, diagnosis, and treatment of various malignants.
Collapse
Affiliation(s)
- Renwei Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Jing Huang
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Yuhao Wei
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Yusha Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Can Lu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Jifeng Liu
- Department of Otolaryngology Head and Neck Surgery/Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- State Key Laboratory of Intelligent Construction and Healthy Operation and Maintenance of Deep Underground Engineering, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| |
Collapse
|
9
|
Qin M, Feng Z, Meng H. Enhanced transcytosis and retention (ETR) effect. Sci Bull (Beijing) 2024; 69:3640-3643. [PMID: 39389865 DOI: 10.1016/j.scib.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Affiliation(s)
- Mengmeng Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Zhenhan Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Meng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
| |
Collapse
|
10
|
Kovshova T, Malinovskaya J, Kotova J, Gorshkova M, Vanchugova L, Osipova N, Melnikov P, Vadekhina V, Nikitin A, Ermolenko Y, Gelperina S. Core-Shell PLGA Nanoparticles: In Vitro Evaluation of System Integrity. Biomolecules 2024; 14:1601. [PMID: 39766308 PMCID: PMC11674307 DOI: 10.3390/biom14121601] [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: 10/28/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The objective of this study was to compare the properties of core-shell nanoparticles with a PLGA core and shells composed of different types of polymers, focusing on their structural integrity. The core PLGA nanoparticles were prepared either through a high-pressure homogenization-solvent evaporation technique or nanoprecipitation, using poloxamer 188 (P188), a copolymer of divinyl ether with maleic anhydride (DIVEMA), and human serum albumin (HSA) as the shell-forming polymers. The shells were formed through adsorption, interfacial embedding, or conjugation. For dual fluorescent labeling, the core- and shell-forming polymers were conjugated with Cyanine5, Cyanine3, and rhodamine B. The nanoparticles had negative zeta potentials and sizes ranging from 100 to 250 nm (measured using DLS) depending on the shell structure and preparation technique. The core-shell structure was confirmed using TEM and fluorescence spectroscopy, with the appearance of FRET phenomena due to the donor-acceptor properties of the labels. All of the shells enhanced the cellular uptake of the nanoparticles in Gl261 murine glioma cells. The integrity of the core-shell structures upon their incubation with the cells was evidenced by intracellular colocalization of the fluorescent labels according to the Manders' colocalization coefficients. This comprehensive approach may be useful for the selection of the optimal preparation method even at the early stages of the core-shell nanoparticle development.
Collapse
Affiliation(s)
- Tatyana Kovshova
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| | - Julia Malinovskaya
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| | - Julia Kotova
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| | - Marina Gorshkova
- Laboratory of Polyelectrolyte Chemistry and Biomedical Polymers, Topchiev Institute of Petrochemical Synthesis, Russian Academy of Sciences, Leninsky Prosp. 29, Moscow 119991, Russia; (M.G.); (L.V.)
| | - Lyudmila Vanchugova
- Laboratory of Polyelectrolyte Chemistry and Biomedical Polymers, Topchiev Institute of Petrochemical Synthesis, Russian Academy of Sciences, Leninsky Prosp. 29, Moscow 119991, Russia; (M.G.); (L.V.)
| | - Nadezhda Osipova
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| | - Pavel Melnikov
- Department of Fundamental and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, Moscow 119034, Russia; (P.M.); (V.V.)
| | - Veronika Vadekhina
- Department of Fundamental and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, Moscow 119034, Russia; (P.M.); (V.V.)
| | - Alexey Nikitin
- Laboratory of Biomedical Nanomaterials, National University of Science and Technology (MISIS), Leninsky Prosp. 4, Moscow 119049, Russia;
| | - Yulia Ermolenko
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| | - Svetlana Gelperina
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| |
Collapse
|
11
|
Cooley M, Wegierak D, Perera R, Abenojar E, Nittayacharn P, Berg FM, Kim Y, Kolios MC, Exner AA. Assessing Therapeutic Nanoparticle Accumulation in Tumors Using Nanobubble-Based Contrast-Enhanced Ultrasound Imaging. ACS NANO 2024; 18:33181-33196. [PMID: 39566912 PMCID: PMC11619768 DOI: 10.1021/acsnano.4c11805] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/22/2024]
Abstract
This study explores the challenges associated with nanoparticle-based drug delivery to the tumor parenchyma, focusing on the widely utilized enhanced permeability and retention effect (EPR). While EPR has been a key strategy, its inconsistent clinical success lacks clear mechanistic understanding and is hindered by limited tools for studying relevant phenomena. This work introduces an approach that employs multiparametric dynamic contrast-enhanced ultrasound (CEUS) with a nanoscale contrast agent for noninvasive, real-time examination of tumor microenvironment characteristics. We demonstrate that CEUS imaging can: (1) evaluate tumor microenvironment features, (2) be used to help predict the distribution of doxorubicin-loaded liposomes in the tumor parenchyma, and (3) be used to predict nanotherapeutic efficacy. CEUS using nanobubbles (NBs) was carried out in two tumor types of high (LS174T) and low (U87) vascular permeability. LS174T tumors consistently showed significantly different time intensity curve (TIC) parameters, including area under the rising curve (AUCR, 2.7×) and time to peak intensity (TTP, 1.9×) compared to U87 tumors. Crucially, a recently developed decorrelation time (DT) parameter specific to NB CEUS dynamics successfully predicted the distribution of doxorubicin-loaded liposomes within the tumor parenchyma (r = 0.86 ± 0.13). AUCR, TTP, and DT were used to correlate imaging findings to nanotherapeutic response with 100% accuracy in SKOV-3 tumors. These findings suggest that NB-CEUS parameters can effectively discern tumor vascular permeability, serving as a biomarker for identifying tumor characteristics and predicting the responsiveness to nanoparticle-based therapies. The observed differences between LS174T and U87 tumors and the accurate prediction of nanotherapeutic efficacy in SKOV-3 tumors indicate the potential utility of this method in predicting treatment efficacy and evaluating EPR in diseases characterized by pathologically permeable vasculature. Ultimately, this research contributes valuable insights into refining drug delivery strategies and assessing the broader applicability of EPR-based approaches.
Collapse
Affiliation(s)
- Michaela
B. Cooley
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland, Ohio 44106, United States
| | - Dana Wegierak
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland, Ohio 44106, United States
| | - Reshani Perera
- Department
of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Eric Abenojar
- Department
of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Pinunta Nittayacharn
- Department
of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, United States
- Department
of Biomedical Engineering, Faculty of Engineering, Mahidol University, Phutthamonthon, Nakhon Pathom 73170, Thailand
| | - Felipe M. Berg
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland, Ohio 44106, United States
- Hospital
Israelita Albert Einstein, São
Paulo, São Paulo 05652-900, Brazil
| | - Youjoung Kim
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland, Ohio 44106, United States
| | - Michael C. Kolios
- Department
of Physics, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
- Institute
for Biomedical Engineering, Science and Technology (iBEST), A Partnership
between St. Michael’s Hospital, A
Site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Agata A. Exner
- Department
of Biomedical Engineering, Case Western
Reserve University, Cleveland, Ohio 44106, United States
- Department
of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
12
|
May JN, Moss JI, Mueller F, Golombek SK, Biancacci I, Rizzo L, Elshafei AS, Gremse F, Pola R, Pechar M, Etrych T, Becker S, Trautwein C, Bülow RD, Boor P, Knuechel R, von Stillfried S, Storm G, Puri S, Barry ST, Schulz V, Kiessling F, Ashford MB, Lammers T. Histopathological biomarkers for predicting the tumour accumulation of nanomedicines. Nat Biomed Eng 2024; 8:1366-1378. [PMID: 38589466 PMCID: PMC7616664 DOI: 10.1038/s41551-024-01197-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/08/2024] [Indexed: 04/10/2024]
Abstract
The clinical prospects of cancer nanomedicines depend on effective patient stratification. Here we report the identification of predictive biomarkers of the accumulation of nanomedicines in tumour tissue. By using supervised machine learning on data of the accumulation of nanomedicines in tumour models in mice, we identified the densities of blood vessels and of tumour-associated macrophages as key predictive features. On the basis of these two features, we derived a biomarker score correlating with the concentration of liposomal doxorubicin in tumours and validated it in three syngeneic tumour models in immunocompetent mice and in four cell-line-derived and six patient-derived tumour xenografts in mice. The score effectively discriminated tumours according to the accumulation of nanomedicines (high versus low), with an area under the receiver operating characteristic curve of 0.91. Histopathological assessment of 30 tumour specimens from patients and of 28 corresponding primary tumour biopsies confirmed the score's effectiveness in predicting the tumour accumulation of liposomal doxorubicin. Biomarkers of the tumour accumulation of nanomedicines may aid the stratification of patients in clinical trials of cancer nanomedicines.
Collapse
Affiliation(s)
- Jan-Niklas May
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, Aachen, Germany
| | - Jennifer I Moss
- Early TDE Discovery, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Florian Mueller
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, Aachen, Germany
| | - Susanne K Golombek
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, Aachen, Germany
| | - Ilaria Biancacci
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, Aachen, Germany
| | - Larissa Rizzo
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, Aachen, Germany
| | - Asmaa Said Elshafei
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, Aachen, Germany
| | - Felix Gremse
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, Aachen, Germany
- Gremse-IT GmbH, Aachen, Germany
| | - Robert Pola
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Michal Pechar
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Svea Becker
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Trautwein
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Aachen, Germany
| | - Roman D Bülow
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Ruth Knuechel
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Saskia von Stillfried
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Gert Storm
- Department of Pharmaceutics, Utrecht University, Utrecht, the Netherlands
- Department of Biomaterials, Science and Technology, University of Twente, Enschede, the Netherlands
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sanyogitta Puri
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | - Simon T Barry
- Early TDE Discovery, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Volkmar Schulz
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, Aachen, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Aachen, Germany
- Physics Institute III B, RWTH Aachen University, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Aachen, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Aachen, Germany
| | - Marianne B Ashford
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, Aachen, Germany.
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Aachen, Germany.
| |
Collapse
|
13
|
Habeeb M, Vengateswaran HT, Tripathi AK, Kumbhar ST, You HW, Hariyadi. Enhancing biomedical imaging: the role of nanoparticle-based contrast agents. Biomed Microdevices 2024; 26:42. [PMID: 39441423 DOI: 10.1007/s10544-024-00725-y] [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] [Accepted: 09/12/2024] [Indexed: 10/25/2024]
Abstract
Biomedical imaging plays a critical role in early detection, precise diagnosis, treatment planning, and monitoring responses, but traditional methods encounter challenges such as limited sensitivity, specificity, and inability to monitor therapeutic responses due to factors like short circulation half-life and potential toxicity. Nanoparticles are revolutionizing biomedical imaging as contrast agents across modalities like computed tomography (CT), optical, magnetic resonance imaging (MRI), and ultrasound, exploiting unique attributes such as those of metal-based, polymeric, and lipid nanoparticles. They shield imaging agents from immune clearance, extending circulation time, and enhancing bioavailability at tumor sites. This results in improved imaging sensitivity. The study highlights advancements in multifunctional nanoparticles for targeted imaging, tackling concerns regarding toxicity and biocompatibility. Critically evaluating conventional contrast agents, emphasizes the shortcomings that nanoparticles aim to overcome. This review provides insight into the current status of nanoparticle-based contrast agents, illuminating their potential to reshape therapeutic monitoring and precision diagnostics.
Collapse
Affiliation(s)
- Mohammad Habeeb
- Department of Pharmaceutics, Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India.
| | - Hariharan Thirumalai Vengateswaran
- Department of Pharmaceutics, Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India.
| | - Arpan Kumar Tripathi
- Department of Pharmacology. KIPS, Shri Shankaracharya Professional University Bhilai, Chhattisgarh, 490020, India
| | - Smita Tukaram Kumbhar
- Department of Pharmaceutical Chemistry, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, Maharashtra, 423603, India
| | - Huay Woon You
- Pusat PERMATA@Pintar Negara, Universiti Kebangsaan Malaysia, 43600, Bangi, Malaysia
| | - Hariyadi
- Department of Electrical Engineering, Muhammadiyah University of West Sumatera, Kota Padang, 26181, Indonesia
| |
Collapse
|
14
|
Jung W, Asaduddin M, Yoo D, Lee DY, Son Y, Kim D, Keum H, Lee J, Park SH, Jon S. Noninvasive ROS imaging and drug delivery monitoring in the tumor microenvironment. Biomaterials 2024; 310:122633. [PMID: 38810387 DOI: 10.1016/j.biomaterials.2024.122633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024]
Abstract
Reactive oxygen species (ROS) that are overproduced in certain tumors can be considered an indicator of oxidative stress levels in the tissue. Here, we report a magnetic resonance imaging (MRI)-based probe capable of detecting ROS levels in the tumor microenvironment (TME) using ROS-responsive manganese ion (Mn2+)-chelated, biotinylated bilirubin nanoparticles (Mn@bt-BRNPs). These nanoparticles are disrupted in the presence of ROS, resulting in the release of free Mn2+, which induces T1-weighted MRI signal enhancement. Mn@BRNPs show more rapid and greater MRI signal enhancement in high ROS-producing A549 lung carcinoma cells compared with low ROS-producing DU145 prostate cancer cells. A pseudo three-compartment model devised for the ROS-reactive MRI probe enables mapping of the distribution and concentration of ROS within the tumor. Furthermore, doxorubicin-loaded, cancer-targeting ligand biotin-conjugated Dox/Mn@bt-BRNPs show considerable accumulation in A549 tumors and also effectively inhibit tumor growth without causing body weight loss, suggesting their usefulness as a new theranostic agent. Collectively, these findings suggest that Mn@bt-BRNPs could be used as an imaging probe capable of detecting ROS levels and monitoring drug delivery in the TME with potential applicability to other inflammatory diseases.
Collapse
Affiliation(s)
- Wonsik Jung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Muhammad Asaduddin
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Dohyun Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Dong Yun Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Seoul, 05505, Republic of Korea
| | - Youngju Son
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Dohyeon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Hyeongseop Keum
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Jungun Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Sung-Hong Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| | - Sangyong Jon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
15
|
Kopeček J. Hydrophilic biomaterials: From crosslinked and self-assembled hydrogels to polymer-drug conjugates and drug-free macromolecular therapeutics. J Control Release 2024; 373:1-22. [PMID: 38734315 PMCID: PMC11384549 DOI: 10.1016/j.jconrel.2024.05.012] [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: 03/15/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024]
Abstract
This "Magnum Opus" accentuates my lifelong belief that the future of science is in the interdisciplinary approach to hypotheses formulation and problem solving. Inspired by the invention of hydrogels and soft contact lenses by my mentors, my six decades of research have continuously proceeded from the synthesis of biocompatible hydrogels to the development of polymer-drug conjugates, then generation of drug-free macromolecular therapeutics (DFMT) and finally to multi-antigen T cell hybridizers (MATCH). This interdisciplinary journey was inspiring; the lifetime feeling that one is a beginner in some aspects of the research is a driving force that keeps the enthusiasm high. Also, I wanted to illustrate that systematic research in one wide area can be a life-time effort without the need to jump to areas that are temporarily en-vogue. In addition to generating general scientific knowledge, hydrogels from my laboratory have been transferred to the clinic, polymer-drug conjugates to clinical trials, and drug-free macromolecular systems have an excellent potential for personalizing patient therapies. There is a limit to life but no limit to imagination. I anticipate that systematic basic research will contribute to the expansion of our knowledge and create a foundation for the design of new paradigms based on the comprehension of mechanisms of physiological processes. The emerging novel platform technologies in biomaterial-based devices and implants as well as in personalized nanomedicines will ultimately impact clinical practice.
Collapse
Affiliation(s)
- Jindřich Kopeček
- Center for Controlled Chemical Delivery, Department of Molecular Pharmaceutics, Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
16
|
Zhang Y, Chen X, Hu B, Zou B, Xu Y. Advancements in nanomedicine delivery systems: unraveling immune regulation strategies for tumor immunotherapy. Nanomedicine (Lond) 2024; 19:1821-1840. [PMID: 39011582 PMCID: PMC11418288 DOI: 10.1080/17435889.2024.2374230] [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: 03/27/2024] [Accepted: 06/26/2024] [Indexed: 07/17/2024] Open
Abstract
This review highlights the significant role of nanodrug delivery systems (NDDS) in enhancing the efficacy of tumor immunotherapy. Focusing on the integration of NDDS with immune regulation strategies, it explores their transformative impacts on the tumor microenvironment and immune response dynamics. Key advancements include the optimization of drug delivery through NDDS, targeting mechanisms like immune checkpoint blockade and modulating the immunosuppressive tumor environment. Despite the progress, challenges such as limited clinical efficacy and complex manufacturing processes persist. The review emphasizes the need for further research to optimize these systems, potentially revolutionizing cancer treatment by improving delivery efficiency, reducing toxicity and overcoming immune resistance.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Xi Chen
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Binbin Hu
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Bingwen Zou
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Yong Xu
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| |
Collapse
|
17
|
Pan Y, Cheng J, Zhu Y, Zhang J, Fan W, Chen X. Immunological nanomaterials to combat cancer metastasis. Chem Soc Rev 2024; 53:6399-6444. [PMID: 38745455 DOI: 10.1039/d2cs00968d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Metastasis causes greater than 90% of cancer-associated deaths, presenting huge challenges for detection and efficient treatment of cancer due to its high heterogeneity and widespread dissemination to various organs. Therefore, it is imperative to combat cancer metastasis, which is the key to achieving complete cancer eradication. Immunotherapy as a systemic approach has shown promising potential to combat metastasis. However, current clinical immunotherapies are not effective for all patients or all types of cancer metastases owing to insufficient immune responses. In recent years, immunological nanomaterials with intrinsic immunogenicity or immunomodulatory agents with efficient loading have been shown to enhance immune responses to eliminate metastasis. In this review, we would like to summarize various types of immunological nanomaterials against metastasis. Moreover, this review will summarize a series of immunological nanomaterial-mediated immunotherapy strategies to combat metastasis, including immunogenic cell death, regulation of chemokines and cytokines, improving the immunosuppressive tumour microenvironment, activation of the STING pathway, enhancing cytotoxic natural killer cell activity, enhancing antigen presentation of dendritic cells, and enhancing chimeric antigen receptor T cell therapy. Furthermore, the synergistic anti-metastasis strategies based on the combinational use of immunotherapy and other therapeutic modalities will also be introduced. In addition, the nanomaterial-mediated imaging techniques (e.g., optical imaging, magnetic resonance imaging, computed tomography, photoacoustic imaging, surface-enhanced Raman scattering, radionuclide imaging, etc.) for detecting metastasis and monitoring anti-metastasis efficacy are also summarized. Finally, the current challenges and future prospects of immunological nanomaterial-based anti-metastasis are also elucidated with the intention to accelerate its clinical translation.
Collapse
Affiliation(s)
- Yuanbo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
- 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 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Junjie Cheng
- Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
- 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 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian, China.
- 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 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 211198, 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 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
| |
Collapse
|
18
|
Liu J, Cabral H, Mi P. Nanocarriers address intracellular barriers for efficient drug delivery, overcoming drug resistance, subcellular targeting and controlled release. Adv Drug Deliv Rev 2024; 207:115239. [PMID: 38437916 DOI: 10.1016/j.addr.2024.115239] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024]
Abstract
The cellular barriers are major bottlenecks for bioactive compounds entering into cells to accomplish their biological functions, which limits their biomedical applications. Nanocarriers have demonstrated high potential and benefits for encapsulating bioactive compounds and efficiently delivering them into target cells by overcoming a cascade of intracellular barriers to achieve desirable therapeutic and diagnostic effects. In this review, we introduce the cellular barriers ahead of drug delivery and nanocarriers, as well as summarize recent advances and strategies of nanocarriers for increasing internalization with cells, promoting intracellular trafficking, overcoming drug resistance, targeting subcellular locations and controlled drug release. Lastly, the future perspectives of nanocarriers for intracellular drug delivery are discussed, which mainly focus on potential challenges and future directions. Our review presents an overview of intracellular drug delivery by nanocarriers, which may encourage the future development of nanocarriers for efficient and precision drug delivery into a wide range of cells and subcellular targets.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Peng Mi
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
19
|
Nomani A, Yousefi S, Sargsyan D, Hatefi A. A quantitative MRI-based approach to estimate the permeation and retention of nanomedicines in tumors. J Control Release 2024; 368:728-739. [PMID: 38493951 PMCID: PMC11412736 DOI: 10.1016/j.jconrel.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Despite the potential of the enhanced permeability and retention (EPR) effect in tumor passive targeting, many nanotherapeutics have failed to produce meaningful clinical outcomes due to the variable and challenging nature of the tumor microenvironment (TME) and EPR effect. This EPR variability across tumors and inconsistent translation of nanomedicines from preclinical to clinical settings necessitates a reliable method to assess its presence in individual tumors. This study aimed to develop a reliable and non-invasive approach to estimate the EPR effect in tumors using a clinically compatible quantitative magnetic resonance imaging (qMRI) technique combined with a nano-sized MRI contrast agent. A quantitative MR imaging was developed using a dynamic contrast-enhanced (DCE) MRI protocol. Then, the permeability and retention of the nano-sized MRI contrast agent were evaluated in three different ovarian xenograft tumor models. Results showed significant differences in EPR effects among the tumor models, with tumor growth influencing the calculated parameters of permeability (Ktrans) and retention (Ve) based on Tofts pharmacokinetic (PK) modeling. Our data indicate that the developed quantitative DCE-MRI method, combined with the Tofts PK modeling, provides a robust and non-invasive approach to screen tumors for their responsiveness to nanotherapeutics. These results imply that the developed qMRI method can be beneficial for personalized cancer treatments by ensuring that nanotherapeutics are administered only to patients with tumors showing sufficient EPR levels.
Collapse
Affiliation(s)
- Alireza Nomani
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA.
| | - Siavash Yousefi
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Davit Sargsyan
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA
| | - Arash Hatefi
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA; Cancer Pharmacology Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA.
| |
Collapse
|
20
|
Sarkar B, Arlauckas SP, Cuccarese MF, Garris CS, Weissleder R, Rodell CB. Host-functionalization of macrin nanoparticles to enable drug loading and control tumor-associated macrophage phenotype. Front Immunol 2024; 15:1331480. [PMID: 38545103 PMCID: PMC10965546 DOI: 10.3389/fimmu.2024.1331480] [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: 11/01/2023] [Accepted: 02/26/2024] [Indexed: 04/09/2024] Open
Abstract
Macrophages are critical regulators of the tumor microenvironment and often present an immuno-suppressive phenotype, supporting tumor growth and immune evasion. Promoting a robust pro-inflammatory macrophage phenotype has emerged as a therapeutic modality that supports tumor clearance, including through synergy with immune checkpoint therapies. Polyglucose nanoparticles (macrins), which possess high macrophage affinity, are useful vehicles for delivering drugs to macrophages, potentially altering their phenotype. Here, we examine the potential of functionalized macrins, synthesized by crosslinking carboxymethyl dextran with L-lysine, as effective carriers of immuno-stimulatory drugs to tumor-associated macrophages (TAMs). Azide groups incorporated during particle synthesis provided a handle for click-coupling of propargyl-modified β-cyclodextrin to macrins under mild conditions. Fluorescence-based competitive binding assays revealed the ability of β-cyclodextrin to non-covalently bind to hydrophobic immuno-stimulatory drug candidates (Keq ~ 103 M-1), enabling drug loading within nanoparticles. Furthermore, transcriptional profiles of macrophages indicated robust pro-inflammatory reprogramming (elevated Nos2 and Il12; suppressed Arg1 and Mrc1 expression levels) for a subset of these immuno-stimulatory agents (UNC2025 and R848). Loading of R848 into the modified macrins improved the drug's effect on primary murine macrophages by three-fold in vitro. Intravital microscopy in IL-12-eYFP reporter mice (24 h post-injection) revealed a two-fold enhancement in mean YFP fluorescence intensity in macrophages targeted with R848-loaded macrins, relative to vehicle controls, validating the desired pro-inflammatory reprogramming of TAMs in vivo by cell-targeted drug delivery. Finally, in an intradermal MC38 tumor model, cyclodextrin-modified macrin NPs loaded with immunostimulatory drugs significantly reduced tumor growth. Therefore, efficient and effective repolarization of tumor-associated macrophages to an M1-like phenotype-via drug-loaded macrins-inhibits tumor growth and may be useful as an adjuvant to existing immune checkpoint therapies.
Collapse
Affiliation(s)
- Biplab Sarkar
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - Sean P. Arlauckas
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
| | - Michael F. Cuccarese
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
| | - Christopher S. Garris
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
- Department of Pathology, Harvard Medical School, Boston, MA, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| | - Christopher B. Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
21
|
Cooley MB, Wegierak D, Exner AA. Using imaging modalities to predict nanoparticle distribution and treatment efficacy in solid tumors: The growing role of ultrasound. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1957. [PMID: 38558290 PMCID: PMC11006412 DOI: 10.1002/wnan.1957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 12/22/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
Nanomedicine in oncology has not had the success in clinical impact that was anticipated in the early stages of the field's development. Ideally, nanomedicines selectively accumulate in tumor tissue and reduce systemic side effects compared to traditional chemotherapeutics. However, this has been more successful in preclinical animal models than in humans. The causes of this failure to translate may be related to the intra- and inter-patient heterogeneity of the tumor microenvironment. Predicting whether a patient will respond positively to treatment prior to its initiation, through evaluation of characteristics like nanoparticle extravasation and retention potential in the tumor, may be a way to improve nanomedicine success rate. While there are many potential strategies to accomplish this, prediction and patient stratification via noninvasive medical imaging may be the most efficient and specific strategy. There have been some preclinical and clinical advances in this area using MRI, CT, PET, and other modalities. An alternative approach that has not been studied as extensively is biomedical ultrasound, including techniques such as multiparametric contrast-enhanced ultrasound (mpCEUS), doppler, elastography, and super-resolution processing. Ultrasound is safe, inexpensive, noninvasive, and capable of imaging the entire tumor with high temporal and spatial resolution. In this work, we summarize the in vivo imaging tools that have been used to predict nanoparticle distribution and treatment efficacy in oncology. We emphasize ultrasound imaging and the recent developments in the field concerning CEUS. The successful implementation of an imaging strategy for prediction of nanoparticle accumulation in tumors could lead to increased clinical translation of nanomedicines, and subsequently, improved patient outcomes. This article is categorized under: Diagnostic Tools In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery Emerging Technologies.
Collapse
Affiliation(s)
- Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana Wegierak
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Radiology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| |
Collapse
|
22
|
Sofias AM, Guo B, Xu J, Lammers T. Image-guided drug delivery: Biomedical and imaging advances. Adv Drug Deliv Rev 2024; 206:115187. [PMID: 38272184 DOI: 10.1016/j.addr.2024.115187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Affiliation(s)
- Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China.
| | - Jian Xu
- Institute of Low-Dimensional Materials Genome Initiative, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
23
|
Long M, Li Y, He H, Gu N. The Story of Ferumoxytol: Synthesis Production, Current Clinical Applications, and Therapeutic Potential. Adv Healthc Mater 2024; 13:e2302773. [PMID: 37931150 DOI: 10.1002/adhm.202302773] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Ferumoxytol, approved by the U.S. Food and Drug Administration in 2009, is one of the intravenous iron oxide nanoparticles authorized for the treatment of iron deficiency in chronic kidney disease and end-stage renal disease. With its exceptional magnetic properties, catalytic activity, and immune activity, as well as good biocompatibility and safety, ferumoxytol has gained significant recognition in various biomedical diagnoses and treatments. Unlike most existing reviews on this topic, this review primarily focuses on the recent clinical and preclinical advances of ferumoxytol in disease treatment, spanning anemia, cancer, infectious inflammatory diseases, regenerative medicine application, magnetic stimulation for neural modulation, etc. Additionally, the newly discovered mechanisms associated with the biological effects of ferumoxytol are discussed, including its magnetic, catalytic, and immunomodulatory properties. Finally, the summary and future prospects concerning the treatment and application of ferumoxytol-based nanotherapeutics are presented.
Collapse
Affiliation(s)
- Mengmeng Long
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Yan Li
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Hongliang He
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- Medical School, Nanjing University, Nanjing, 210008, P. R. China
| |
Collapse
|
24
|
Deng H, Yang X, Wang H, Gao M, Zhang Y, Liu R, Xu H, Zhang W. Tailoring the surface charges of iron-crosslinked dextran nanogels towards improved tumor-associated macrophage targeting. Carbohydr Polym 2024; 325:121585. [PMID: 38008480 DOI: 10.1016/j.carbpol.2023.121585] [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: 09/09/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/28/2023]
Abstract
Tumor-associated macrophages (TAMs) have emerged as therapeutic interests in cancer nanomedicine because TAMs play a pivotal role in the immune microenvironment of solid tumors. Dextran and its derived nanocarriers are among the most promising nanomaterials for TAM targeting due to their intrinsic affinities towards macrophages. Various dextran-based nanomaterials have been developed to image TAMs. However, the effects of physiochemical properties especially for surface charges of dextran nanomaterials on TAM-targeting efficacy were ambiguous in literature. To figure out the surface charge effects on TAM targeting, here we developed a facile non-covalent self-assembly strategy to construct oppositely charged dextran nanogels (NGs) utilizing the coordination interaction of ferric ions, chlorine e6 (Ce6) dye and three dextran derivatives, diethylaminoethyl-, sulfate sodium- and carboxymethyl-dextran. The acquired dextran NGs exhibit different charges but similar hydrodynamic size, Ce6 loading and mechanical stiffness, which enables a side-by-side comparison of the effects of NG surface charges on TAM targeting monitored by the Ce6 fluorescence imaging. Compared with negative NGs, the positive NG clearly displays a superior TAM targeting in murine breast cancer model. This study identifies that positively charged dextran NG could be a promising approach to better engineer nanomedicine towards an improved TAM targeting.
Collapse
Affiliation(s)
- Hong Deng
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China; Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Xue Yang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Huimin Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China; Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Menghan Gao
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China; Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Yiyi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China; Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Runmeng Liu
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China; Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Haiyan Xu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China.
| | - Weiqi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China; Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China.
| |
Collapse
|
25
|
Patel H, Li J, Bo L, Mehta R, Ashby CR, Wang S, Cai W, Chen ZS. Nanotechnology-based delivery systems to overcome drug resistance in cancer. MEDICAL REVIEW (2021) 2024; 4:5-30. [PMID: 38515777 PMCID: PMC10954245 DOI: 10.1515/mr-2023-0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/24/2024] [Indexed: 03/23/2024]
Abstract
Cancer nanomedicine is defined as the application of nanotechnology and nanomaterials for the formulation of cancer therapeutics that can overcome the impediments and restrictions of traditional chemotherapeutics. Multidrug resistance (MDR) in cancer cells can be defined as a decrease or abrogation in the efficacy of anticancer drugs that have different molecular structures and mechanisms of action and is one of the primary causes of therapeutic failure. There have been successes in the development of cancer nanomedicine to overcome MDR; however, relatively few of these formulations have been approved by the United States Food and Drug Administration for the treatment of cancer. This is primarily due to the paucity of knowledge about nanotechnology and the fundamental biology of cancer cells. Here, we discuss the advances, types of nanomedicines, and the challenges regarding the translation of in vitro to in vivo results and their relevance to effective therapies.
Collapse
Affiliation(s)
- Harsh Patel
- College of Pharmacy and Health Sciences, St. John’s University, New York, NY, USA
| | - Jiaxin Li
- College of Pharmacy and Health Sciences, St. John’s University, New York, NY, USA
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, Hunan Province, China
| | - Letao Bo
- College of Pharmacy and Health Sciences, St. John’s University, New York, NY, USA
| | - Riddhi Mehta
- St. John’s College of Liberal Arts and Sciences, St. John’s University, New York, NY, USA
| | - Charles R. Ashby
- College of Pharmacy and Health Sciences, St. John’s University, New York, NY, USA
| | - Shanzhi Wang
- College of Pharmacy and Health Sciences, St. John’s University, New York, NY, USA
| | - Wei Cai
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, Hunan Province, China
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John’s University, New York, NY, USA
| |
Collapse
|
26
|
Morales CS, Grodzinski P. Current landscape of treating different cancers using nanomedicines: Trends and perspectives. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1927. [PMID: 37706362 DOI: 10.1002/wnan.1927] [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: 07/28/2023] [Accepted: 08/21/2023] [Indexed: 09/15/2023]
Abstract
The efforts to use novel nanotechnologies in medicine and cancer have been widespread. In order to understand better the focus areas of cancer nanomedicine research to date, we conducted a survey of nanomedicine developmental and clinical research in conjunction with treatment of various cancers. The survey has been performed based on number of publications, rate of citations, entry into clinical trials, and funding rates by the National Cancer Institute. Our survey indicates that breast and brain cancers are the most and one of the least studied by nanotechnology researchers, respectively. Breast cancer nano-therapies seem to also be most likely to achieve clinical translation as the number of publications produced, amount of funding, total citations, and clinical trials (active and completed) are the highest when compared with research in other cancers. Brain cancer, despite its low survival, has capture much less attention of nanomedicine research community as survey indicated, although nanotechnology can offer novel approaches which can address brain cancer challenges. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Carolina Salvador Morales
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Piotr Grodzinski
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
27
|
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
|
28
|
Cooley MB, Wegierak D, Perera R, Abenojar EC, Nittayacharn PA, Berg FM, Kim Y, Kolios MC, Exner AA. Assessing Tumor Microenvironment Characteristics and Stratifying EPR with a Nanobubble Companion Nanoparticle via Contrast-Enhanced Ultrasound Imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567934. [PMID: 38045236 PMCID: PMC10690218 DOI: 10.1101/2023.11.20.567934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
The tumor microenvironment is characterized by dysfunctional endothelial cells, resulting in heightened vascular permeability. Many nanoparticle-based drug delivery systems attempt to use this enhanced permeability combined with impaired lymphatic drainage (a concept known as the 'enhanced permeability and retention effect' or EPR effect) as the primary strategy for drug delivery, but this has not proven to be as clinically effective as anticipated. The specific mechanisms behind the inconsistent clinical outcomes of nanotherapeutics have not been clearly articulated, and the field has been hampered by a lack of accessible tools to study EPR-associated phenomena in clinically relevant scenarios. While medical imaging has tremendous potential to contribute to this area, it has not been broadly explored. This work examines, for the first time, the use of multiparametric dynamic contrast-enhanced ultrasound (CEUS) with a novel nanoscale contrast agent to examine tumor microenvironment characteristics noninvasively and in real-time. We demonstrate that CEUS imaging can: (1) evaluate tumor microenvironment features and (2) be used to help predict the distribution of doxorubicin-loaded liposomes in the tumor parenchyma. CEUS using nanobubbles (NBs) was carried out in two tumor types of high (LS174T) and low (U87) vascular permeability, and time-intensity curve (TIC) parameters were evaluated in both models prior to injection of doxorubicin liposomes. Consistently, LS174T tumors showed significantly different TIC parameters, including area under the rising curve (2.7x), time to peak intensity (1.9x) and decorrelation time (DT, 1.9x) compared to U87 tumors. Importantly, the DT parameter successfully predicted tumoral nanoparticle distribution (r = 0.86 ± 0.13). Ultimately, substantial differences in NB-CEUS generated parameters between LS174T and U87 tumors suggest that this method may be useful in determining tumor vascular permeability and could be used as a biomarker for identifying tumor characteristics and predicting sensitivity to nanoparticle-based therapies. These findings could ultimately be applied to predicting treatment efficacy and to evaluating EPR in other diseases with pathologically permeable vasculature.
Collapse
|
29
|
Wang C, Zhang Y. Current Application of Nanoparticle Drug Delivery Systems to the Treatment of Anaplastic Thyroid Carcinomas. Int J Nanomedicine 2023; 18:6037-6058. [PMID: 37904863 PMCID: PMC10613415 DOI: 10.2147/ijn.s429629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/18/2023] [Indexed: 11/01/2023] Open
Abstract
Anaplastic thyroid carcinomas (ATCs) are a rare subtype of thyroid cancers with a low incidence but extremely high invasiveness and fatality. The treatment of ATCs is very challenging, and currently, a comprehensive individualized therapeutic strategy involving surgery, radiotherapy (RT), chemotherapy, BRAF/MEK inhibitors (BRAFi/MEKi) and immunotherapy is preferred. For ATC patients in stage IVA/IVB, a surgery-based comprehensive strategy may provide survival benefits. Unfortunately, ATC patients in IVC stage barely get benefits from the current treatment. Recently, nanoparticle delivery of siRNAs, targeted drugs, cytotoxic drugs, photosensitizers and other agents is considered as a promising anti-cancer treatment. Nanoparticle drug delivery systems have been mainly explored in the treatment of differentiated thyroid cancer (DTC). With the rapid development of drug delivery techniques and nanomaterials, using hybrid nanoparticles as the drug carrier to deliver siRNAs, targeted drugs, immune drugs, chemotherapy drugs and phototherapy drugs to ATC patients have become a hot research field. This review aims to describe latest findings of nanoparticle drug delivery systems in the treatment of ATCs, thus providing references for the further analyses.
Collapse
Affiliation(s)
- Chonggao Wang
- Department of Thyroid Surgery, Nanjing Hospital of Chinese Medicine, Nanjing, 210001, People’s Republic of China
- School of Medicine, Southeast University, Nanjing, 210001, People’s Republic of China
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210009, People’s Republic of China
| |
Collapse
|
30
|
Ma X, Mao M, He J, Liang C, Xie HY. Nanoprobe-based molecular imaging for tumor stratification. Chem Soc Rev 2023; 52:6447-6496. [PMID: 37615588 DOI: 10.1039/d3cs00063j] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
The responses of patients to tumor therapies vary due to tumor heterogeneity. Tumor stratification has been attracting increasing attention for accurately distinguishing between responders to treatment and non-responders. Nanoprobes with unique physical and chemical properties have great potential for patient stratification. This review begins by describing the features and design principles of nanoprobes that can visualize specific cell types and biomarkers and release inflammatory factors during or before tumor treatment. Then, we focus on the recent advancements in using nanoprobes to stratify various therapeutic modalities, including chemotherapy, radiotherapy (RT), photothermal therapy (PTT), photodynamic therapy (PDT), chemodynamic therapy (CDT), ferroptosis, and immunotherapy. The main challenges and perspectives of nanoprobes in cancer stratification are also discussed to facilitate probe development and clinical applications.
Collapse
Affiliation(s)
- Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Mingchuan Mao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jiaqi He
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing, 100191, P. R. China.
| |
Collapse
|
31
|
Sun S, Wang YH, Gao X, Wang HY, Zhang L, Wang N, Li CM, Xiong SQ. Current perspectives and trends in nanoparticle drug delivery systems in breast cancer: bibliometric analysis and review. Front Bioeng Biotechnol 2023; 11:1253048. [PMID: 37771575 PMCID: PMC10523396 DOI: 10.3389/fbioe.2023.1253048] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/04/2023] [Indexed: 09/30/2023] Open
Abstract
The treatment of breast cancer (BC) is a serious challenge due to its heterogeneous nature, multidrug resistance (MDR), and limited therapeutic options. Nanoparticle-based drug delivery systems (NDDSs) represent a promising tool for overcoming toxicity and chemotherapy drug resistance in BC treatment. No bibliometric studies have yet been published on the research landscape of NDDS-based treatment of BC. In this review, we extracted data from 1,752 articles on NDDS-based treatment of BC published between 2012 and 2022 from the Web of Science Core Collection (WOSCC) database. VOSviewer, CiteSpace, and some online platforms were used for bibliometric analysis and visualization. Publication trends were initially observed: in terms of geographical distribution, China and the United States had the most papers on this subject. The highest contributing institution was Sichuan University. In terms of authorship and co-cited authorship, the most prolific author was Yu Zhang. Furthermore, Qiang Zhang and co-workers have made tremendous achievements in the field of NDDS-based BC treatment. The article titled "Nanomedicine in cancer therapy: challenges, opportunities, and clinical applications" had the most citations. The Journal of Controlled Release was one of the most active publishers in the field. "Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries" was the most cited reference. We also analysed "hot" and cutting-edge research for NDDSs in BC treatment. There were nine topic clusters: "tumour microenvironment," "nanoparticles (drug delivery)," "breast cancer/triple-negative breast cancer," "combination therapy," "drug release (pathway)," "multidrug resistance," "recent advance," "targeted drug delivery", and "cancer nanomedicine." We also reviewed the core themes of research. In summary, this article reviewed the application of NDDSs in the treatment of BC.
Collapse
Affiliation(s)
- Sheng Sun
- Sichuan Integrative Medicine Hospital, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ye-hui Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Gao
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - He-yong Wang
- Sichuan Integrative Medicine Hospital, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Na Wang
- Sichuan Integrative Medicine Hospital, Chengdu, China
| | - Chun-mei Li
- Sichuan Integrative Medicine Hospital, Chengdu, China
| | - Shao-quan Xiong
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
32
|
Zhao DK, Liang J, Huang XY, Shen S, Wang J. Organoids technology for advancing the clinical translation of cancer nanomedicine. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1892. [PMID: 37088100 DOI: 10.1002/wnan.1892] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 04/25/2023]
Abstract
The past decades have witnessed the rapid development and widespread application of nanomedicines in cancer treatment; however, the clinical translation of experimental findings has been low, as evidenced by the low percentage of commercialized nanomedicines. Incomplete understanding of nanomedicine-tumor interactions and inappropriate evaluation models are two important challenges limiting the clinical translation of cancer nanomedicines. Currently, nanomedicine-tumor interaction and therapeutic effects are mainly investigated using cell lines or mouse models, which do not recapitulate the complex tumor microenvironment in human patients. Thus, information obtained from cell lines and mouse models cannot provide adequate guidance for the rational redesign of nanomedicine. Compared with other preclinical models, tumor organoids constructed from patient-derived tumor tissues are superior in retaining the key histopathological, genetic, and phenotypic features of the parent tumor. We speculate that organoid technology would help elucidate nanomedicine-tumor interaction in the tumor microenvironment and guide the design of nanomedicine, making it a reliable tool to accurately predict drug responses in patients with cancer. This review highlighted the advantages of drug delivery systems in cancer treatment, challenges limiting the clinical translation of antitumor nanomedicines, and potential application of patient-derived organoids (PDO) in nanomedicine. We propose that combining organoids and nanotechnology would facilitate the development of safe and effective cancer nanomedicines and accelerate their clinical application. This review discussed the potential translational value of integrative research using organoids and cancer nanomedicine. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Dong-Kun Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Jie Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Xiao-Yi Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, China
| |
Collapse
|
33
|
Jazwinska DE, Kulawiec DG, Zervantonakis IK. Cancer-mesothelial and cancer-macrophage interactions in the ovarian cancer microenvironment. Am J Physiol Cell Physiol 2023; 325:C721-C730. [PMID: 37545408 PMCID: PMC10635648 DOI: 10.1152/ajpcell.00461.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Abstract
The metastatic ovarian cancer microenvironment is characterized by an intricate interaction network between cancer cells and host cells. This complex heterotypic cancer-host cell crosstalk results in an environment that promotes cancer cell metastasis and treatment resistance, leading to poor patient prognosis and survival. In this review, we focus on two host cell types found in the ovarian cancer microenvironment: mesothelial cells and tumor-associated macrophages. Mesothelial cells make up the protective lining of organs in the abdominal cavity. Cancer cells attach and invade through the mesothelial monolayer to form metastatic lesions. Crosstalk between mesothelial and cancer cells can contribute to metastatic progression and chemotherapy resistance. Tumor-associated macrophages are the most abundant immune cell type in the ovarian cancer microenvironment with heterogeneous subpopulations exhibiting protumor or antitumor functions. Macrophage reprogramming toward a protumor or antitumor state can be influenced by chemotherapy and communication with cancer cells, resulting in cancer cell invasion and treatment resistance. A better understanding of cancer-mesothelial and cancer-macrophage crosstalk will uncover biomarkers of metastatic progression and therapeutic targets to restore chemotherapy sensitivity.
Collapse
Affiliation(s)
- Dorota E Jazwinska
- Department of Bioengineering and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Diana G Kulawiec
- Department of Bioengineering and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Ioannis K Zervantonakis
- Department of Bioengineering and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
34
|
Hicks MR, Liu X, Young CS, Saleh K, Ji Y, Jiang J, Emami MR, Mokhonova E, Spencer MJ, Meng H, Pyle AD. Nanoparticles systemically biodistribute to regenerating skeletal muscle in DMD. J Nanobiotechnology 2023; 21:303. [PMID: 37641124 PMCID: PMC10463982 DOI: 10.1186/s12951-023-01994-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 07/09/2023] [Indexed: 08/31/2023] Open
Abstract
Skeletal muscle disease severity can often progress asymmetrically across muscle groups and heterogeneously within tissues. An example is Duchenne Muscular Dystrophy (DMD) in which lack of dystrophin results in devastating skeletal muscle wasting in some muscles whereas others are spared or undergo hypertrophy. An efficient, non-invasive approach to identify sites of asymmetry and degenerative lesions could enable better patient monitoring and therapeutic targeting of disease. In this study, we utilized a versatile intravenously injectable mesoporous silica nanoparticle (MSNP) based nanocarrier system to explore mechanisms of biodistribution in skeletal muscle of mdx mouse models of DMD including wildtype, dystrophic, and severely dystrophic mice. Moreover, MSNPs could be imaged in live mice and whole muscle tissues enabling investigation of how biodistribution is altered by different types of muscle pathology such as inflammation or fibrosis. We found MSNPs were tenfold more likely to aggregate within select mdx muscles relative to wild type, such as gastrocnemius and quadriceps. This was accompanied by decreased biodistribution in off-target organs. We found the greatest factor affecting preferential delivery was the regenerative state of the dystrophic skeletal muscle with the highest MSNP abundance coinciding with the regions showing the highest level of embryonic myosin staining and intramuscular macrophage uptake. To demonstrate, muscle regeneration regulated MSNP distribution, we experimentally induced regeneration using barium chloride which resulted in a threefold increase of intravenously injected MSNPs to sites of regeneration 7 days after injury. These discoveries provide the first evidence that nanoparticles have selective biodistribution to skeletal muscle in DMD to areas of active regeneration and that nanoparticles could enable diagnostic and selective drug delivery in DMD skeletal muscle.
Collapse
Affiliation(s)
- Michael R Hicks
- Department of Microbiology, Immunology and Medical Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Eli and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Los Angeles, CA, USA
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Xiangsheng Liu
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- California Nanosystems Institute at UCLA, Los Angeles, CA, USA
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Courtney S Young
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- MyoGene Bio, San Diego, CA, USA
| | - Kholoud Saleh
- Department of Microbiology, Immunology and Medical Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ying Ji
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jinhong Jiang
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- California Nanosystems Institute at UCLA, Los Angeles, CA, USA
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Michael R Emami
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ekaterina Mokhonova
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Melissa J Spencer
- Eli and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Los Angeles, CA, USA.
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| | - Huan Meng
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- California Nanosystems Institute at UCLA, Los Angeles, CA, USA.
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China.
| | - April D Pyle
- Department of Microbiology, Immunology and Medical Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Eli and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Los Angeles, CA, USA.
| |
Collapse
|
35
|
Liao S, Zhou M, Wang Y, Lu C, Yin B, Zhang Y, Liu H, Yin X, Song G. Emerging biomedical imaging-based companion diagnostics for precision medicine. iScience 2023; 26:107277. [PMID: 37520706 PMCID: PMC10371849 DOI: 10.1016/j.isci.2023.107277] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023] Open
Abstract
The tumor heterogeneity, which leads to individual variations in tumor microenvironments, causes poor prognoses and limits therapeutic response. Emerging technology such as companion diagnostics (CDx) detects biomarkers and monitors therapeutic responses, allowing identification of patients who would benefit most from treatment. However, currently, most US Food and Drug Administration-approved CDx tests are designed to detect biomarkers in vitro and ex vivo, making it difficult to dynamically report variations of targets in vivo. Various medical imaging techniques offer dynamic measurement of tumor heterogeneity and treatment response, complementing CDx tests. Imaging-based companion diagnostics allow for patient stratification for targeted medicines and identification of patient populations benefiting from alternative therapeutic methods. This review summarizes recent developments in molecular imaging for predicting and assessing responses to cancer therapies, as well as the various biomarkers used in imaging-based CDx tests. We hope this review provides informative insights into imaging-based companion diagnostics and advances precision medicine.
Collapse
Affiliation(s)
- Shiyi Liao
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Mengjie Zhou
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Youjuan Wang
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Chang Lu
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Baoli Yin
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Ying Zhang
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Huiyi Liu
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Xia Yin
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Guosheng Song
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
36
|
Fan D, Cao Y, Cao M, Wang Y, Cao Y, Gong T. Nanomedicine in cancer therapy. Signal Transduct Target Ther 2023; 8:293. [PMID: 37544972 PMCID: PMC10404590 DOI: 10.1038/s41392-023-01536-y] [Citation(s) in RCA: 219] [Impact Index Per Article: 109.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/08/2023] Open
Abstract
Cancer remains a highly lethal disease in the world. Currently, either conventional cancer therapies or modern immunotherapies are non-tumor-targeted therapeutic approaches that cannot accurately distinguish malignant cells from healthy ones, giving rise to multiple undesired side effects. Recent advances in nanotechnology, accompanied by our growing understanding of cancer biology and nano-bio interactions, have led to the development of a series of nanocarriers, which aim to improve the therapeutic efficacy while reducing off-target toxicity of the encapsulated anticancer agents through tumor tissue-, cell-, or organelle-specific targeting. However, the vast majority of nanocarriers do not possess hierarchical targeting capability, and their therapeutic indices are often compromised by either poor tumor accumulation, inefficient cellular internalization, or inaccurate subcellular localization. This Review outlines current and prospective strategies in the design of tumor tissue-, cell-, and organelle-targeted cancer nanomedicines, and highlights the latest progress in hierarchical targeting technologies that can dynamically integrate these three different stages of static tumor targeting to maximize therapeutic outcomes. Finally, we briefly discuss the current challenges and future opportunities for the clinical translation of cancer nanomedicines.
Collapse
Affiliation(s)
- Dahua Fan
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China.
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| | - Yongkai Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Meiqun Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Yajun Wang
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China
| | | | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
37
|
Zhang J, Wang S, Zhang D, He X, Wang X, Han H, Qin Y. Nanoparticle-based drug delivery systems to enhance cancer immunotherapy in solid tumors. Front Immunol 2023; 14:1230893. [PMID: 37600822 PMCID: PMC10435760 DOI: 10.3389/fimmu.2023.1230893] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Immunotherapy has developed rapidly in solid tumors, especially in the areas of blocking inhibitory immune checkpoints and adoptive T-cell transfer for immune regulation. Many patients benefit from immunotherapy. However, the response rate of immunotherapy in the overall population are relatively low, which depends on the characteristics of the tumor and individualized patient differences. Moreover, the occurrence of drug resistance and adverse reactions largely limit the development of immunotherapy. Recently, the emergence of nanodrug delivery systems (NDDS) seems to improve the efficacy of immunotherapy by encapsulating drug carriers in nanoparticles to precisely reach the tumor site with high stability and biocompatibility, prolonging the drug cycle of action and greatly reducing the occurrence of toxic side effects. In this paper, we mainly review the advantages of NDDS and the mechanisms that enhance conventional immunotherapy in solid tumors, and summarize the recent advances in NDDS-based therapeutic strategies, which will provide valuable ideas for the development of novel tumor immunotherapy regimen.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Siyuan Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Daidi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin He
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xue Wang
- Academy of Medical Science, School of Basic Medical Science, Zhengzhou University, Zhengzhou, China
| | - Huiqiong Han
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
38
|
Abstract
The host immune system possesses an intrinsic ability to target and kill cancer cells in a specific and adaptable manner that can be further enhanced by cancer immunotherapy, which aims to train the immune system to boost the antitumor immune response. Several different categories of cancer immunotherapy have emerged as new standard cancer therapies in the clinic, including cancer vaccines, immune checkpoint inhibitors, adoptive T cell therapy, and oncolytic virus therapy. Despite the remarkable survival benefit for a subset of patients, the low response rate and immunotoxicity remain the major challenges for current cancer immunotherapy. Over the last few decades, nanomedicine has been intensively investigated with great enthusiasm, leading to marked advancements in nanoparticle platforms and nanoengineering technology. Advances in nanomedicine and immunotherapy have also led to the emergence of a nascent research field of nano-immunotherapy, which aims to realize the full therapeutic potential of immunotherapy with the aid of nanomedicine. In particular, nanocarriers present an exciting opportunity in immuno-oncology to boost the activity, increase specificity, decrease toxicity, and sustain the antitumor efficacy of immunological agents by potentiating immunostimulatory activity and favorably modulating pharmacological properties. This review discusses the potential of nanocarriers for cancer immunotherapy and introduces preclinical studies designed to improve clinical cancer immunotherapy modalities using nanocarrier-based engineering approaches. It also discusses the potential of nanocarriers to address the challenges currently faced by immuno-oncology as well as the challenges for their translation to clinical applications.
Collapse
Affiliation(s)
- Isra Rana
- College of Pharmacy, Chonnam National University, Gwangju, 61186, South Korea
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Jaeeun Oh
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea
| | - Juwon Baig
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea
| | - Jeong Hyun Moon
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea
| | - Sejin Son
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea.
- Department of Biological Sciences and Bioengineering, Inha University/Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon, South Korea.
| | - Jutaek Nam
- College of Pharmacy, Chonnam National University, Gwangju, 61186, South Korea.
| |
Collapse
|
39
|
Soni SS, D'Elia AM, Rodell CB. Control of the post-infarct immune microenvironment through biotherapeutic and biomaterial-based approaches. Drug Deliv Transl Res 2023; 13:1983-2014. [PMID: 36763330 PMCID: PMC9913034 DOI: 10.1007/s13346-023-01290-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2023] [Indexed: 02/11/2023]
Abstract
Ischemic heart failure (IHF) is a leading cause of morbidity and mortality worldwide, for which heart transplantation remains the only definitive treatment. IHF manifests from myocardial infarction (MI) that initiates tissue remodeling processes, mediated by mechanical changes in the tissue (loss of contractility, softening of the myocardium) that are interdependent with cellular mechanisms (cardiomyocyte death, inflammatory response). The early remodeling phase is characterized by robust inflammation that is necessary for tissue debridement and the initiation of repair processes. While later transition toward an immunoregenerative function is desirable, functional reorientation from an inflammatory to reparatory environment is often lacking, trapping the heart in a chronically inflamed state that perpetuates cardiomyocyte death, ventricular dilatation, excess fibrosis, and progressive IHF. Therapies can redirect the immune microenvironment, including biotherapeutic and biomaterial-based approaches. In this review, we outline these existing approaches, with a particular focus on the immunomodulatory effects of therapeutics (small molecule drugs, biomolecules, and cell or cell-derived products). Cardioprotective strategies, often focusing on immunosuppression, have shown promise in pre-clinical and clinical trials. However, immunoregenerative therapies are emerging that often benefit from exacerbating early inflammation. Biomaterials can be used to enhance these therapies as a result of their intrinsic immunomodulatory properties, parallel mechanisms of action (e.g., mechanical restraint), or by enabling cell or tissue-targeted delivery. We further discuss translatability and the continued progress of technologies and procedures that contribute to the bench-to-bedside development of these critically needed treatments.
Collapse
Affiliation(s)
- Shreya S Soni
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Arielle M D'Elia
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Christopher B Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA.
| |
Collapse
|
40
|
Yan X, Li S, Yan H, Yu C, Liu F. IONPs-Based Medical Imaging in Cancer Care: Moving Beyond Traditional Diagnosis and Therapeutic Assessment. Int J Nanomedicine 2023; 18:1741-1763. [PMID: 37034271 PMCID: PMC10075272 DOI: 10.2147/ijn.s399047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 03/14/2023] [Indexed: 04/03/2023] Open
Abstract
Cancer-related burden of morbidity and mortality is rapidly rising worldwide. Medical imaging plays an important role in every phase of cancer management, including diagnosis, staging, treatment planning and evaluation. Iron oxide nanoparticles (IONPs) could serve as contrast agents or labeling agents to enhance the identification and visualization of pathological tissues as well as target cells. Multimodal or multifunctional imaging can be easily acquired by modifying IONPs with other imaging agents or functional groups, allowing the accessibility of combined imaging techniques and providing more comprehensive information for cancer care. To date, IONPs-enhanced medical imaging has gained intensive application in early diagnosis, monitoring treatment as well as guiding radio-frequency ablation, sentinel lymph node dissection, radiotherapy and hyperthermia therapy. Besides, IONPs mediated imaging is also capable of promoting the development of anti-cancer nanomedicines through identifying patients potentially sensitive to nanotherapeutics. Based on versatile imaging modes and application fields, this review highlights and summarizes recent research advances of IONPs-based medical imaging in cancer management. Besides, currently existing challenges are also discussed to provide perspectives and advices for the future development of IONPs-based imaging in cancer management.
Collapse
Affiliation(s)
- Xiaolin Yan
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong Province, People’s Republic of China
| | - Shanshan Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong Province, People’s Republic of China
| | - Haiyin Yan
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong Province, People’s Republic of China
| | - Chungang Yu
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong Province, People’s Republic of China
| | - Fengxi Liu
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong Province, People’s Republic of China
- Correspondence: Fengxi Liu, Tel +86 0531-89269594, Email
| |
Collapse
|
41
|
Ravi H, Arias-Lorza AM, Costello JR, Han HS, Jeong DK, Klinz SG, Sachdev JC, Korn RL, Raghunand N. Pretherapy Ferumoxytol-enhanced MRI to Predict Response to Liposomal Irinotecan in Metastatic Breast Cancer. Radiol Imaging Cancer 2023; 5:e220022. [PMID: 36734848 PMCID: PMC10077095 DOI: 10.1148/rycan.220022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Purpose To investigate ferumoxytol (FMX)-enhanced MRI as a pretreatment predictor of response to liposomal irinotecan (nal-IRI) for thoracoabdominal and brain metastases in women with metastatic breast cancer (mBC). Materials and Methods In this phase 1 expansion trial (ClinicalTrials.gov identifier, NCT01770353; 27 participants), 49 thoracoabdominal (19 participants; mean age, 48 years ± 11 [SD]) and 19 brain (seven participants; mean age, 54 years ± 8) metastases were analyzed on MR images acquired before, 1-4 hours after, and 16-24 hours after FMX administration. In thoracoabdominal metastases, tumor transverse relaxation rate (R*2) was normalized to the mean R*2 in the spleen (rR*2), and the tumor histogram metric rR*2,N, representing the average of rR*2 in voxels above the nth percentile, was computed. In brain metastases, a novel compartmentation index was derived by applying the MRI signal equation to phantom-calibrated coregistered FMX-enhanced MRI brain scans acquired before, 1-4 hours after, and 16-24 hours after FMX administration. The fraction of voxels with an FMX compartmentation index greater than 1 was computed over the whole tumor (FCIGT1) and from voxels above the 90th percentile R*2 (FCIGT1 R*2,90). Results rR*2,90 computed from pretherapy MRI performed 16-24 hours after FMX administration, without reference to calibration phantoms, predicted response to nal-IRI in thoracoabdominal metastases (accuracy, 74%). rR*2,90 performance was robust to the inclusion of some peritumoral tissue within the tumor region of interest. FCIGT1 R*2,90 provided 79% accuracy on cross-validation in prediction of response in brain metastases. Conclusion This first in-human study focused on mBC suggests that FMX-enhanced MRI biologic markers can be useful for pretherapy prediction of response to nal-IRI in patients with mBC. Keywords: MRI Contrast Agent, MRI, Breast, Head/Neck, Tumor Response, Experimental Investigations, Brain/Brain Stem Clinical trial registration no. NCT01770353 Supplemental material is available for this article. © RSNA, 2023 See also commentary by Daldrup-Link in this issue.
Collapse
Affiliation(s)
- Harshan Ravi
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Andres M Arias-Lorza
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - James R Costello
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Hyo Sook Han
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Daniel K Jeong
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Stephan G Klinz
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Jasgit C Sachdev
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Ronald L Korn
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| | - Natarajan Raghunand
- From the Departments of Cancer Physiology (H.R., A.M.A.L., N.R.), Radiology (J.R.C., D.K.J.), and Breast Oncology (H.S.H.), Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL 33612; Ipsen Bioscience, Cambridge, Mass (S.G.K.); HonorHealth Research Institute, Scottsdale, Ariz (J.C.S.); Imaging Endpoints Core Laboratory, Scottsdale, Ariz (R.L.K.); and Department of Oncologic Sciences, University of South Florida, Tampa, Fla (N.R.)
| |
Collapse
|
42
|
Yang W, Deng C, Shi X, Xu Y, Dai C, Wang H, Bian K, Cui T, Zhang B. Structural and Molecular Fusion MRI Nanoprobe for Differential Diagnosis of Malignant Tumors and Follow-Up Chemodynamic Therapy. ACS NANO 2023; 17:4009-4022. [PMID: 36757738 DOI: 10.1021/acsnano.2c12874] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Enhanced imaging techniques using contrast agents enable high-resolution structural imaging to reveal space-occupying lesions but rarely provide detailed molecular information. To this end, we report a structural and molecular fusion magnetic resonance imaging (MRI) nanoprobe for differential diagnosis between benign and malignant tumors. This fusion nanoprobe, termed FFT NPs, follows a working mechanism involving a T1-/T2-weighted magnetic resonance tuning effect (MRET) between a magnetic Fe3O4 core and a paramagnetic Fe-tannic acid (Fe-TA) shell. The FFT NPs with an "always-on" inert T2 signal provide structural MRI (sMRI) contrast of tumors while affording an activated T1 signal in the presence of ATP, which is overproduced during the rapid growth of malignant tumors to enable molecular MRI (mMRI) of tumor lesions. We propose the use of the ratiometric mMRI:sMRI intensity to assist in the differential diagnosis of malignant 4T1 tumors from benign L929 fibroblast tumors. Furthermore, the dissociated FFT NPs were found to be able to catalyze H2O2 conversion in 4T1 tumors to generate excess reactive oxygen species (ROS) for chemodynamic therapy. The described fusion nanoprobe strategy enables the differential diagnosis of tumors from a combined spatial and molecular perspective with one-stop MRI imaging with potential applications in precision intervention.
Collapse
Affiliation(s)
- Weitao Yang
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
| | - Cuijun Deng
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People's Hospital, Collaborative Innovation Center for Brain Science, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xiudong Shi
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yan Xu
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
| | - Chenyu Dai
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
| | - Hui Wang
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
| | - Kexin Bian
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
| | - Tianming Cui
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
| | - Bingbo Zhang
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
| |
Collapse
|
43
|
Turco V, Pfleiderer K, Hunger J, Horvat NK, Karimian-Jazi K, Schregel K, Fischer M, Brugnara G, Jähne K, Sturm V, Streibel Y, Nguyen D, Altamura S, Agardy DA, Soni SS, Alsasa A, Bunse T, Schlesner M, Muckenthaler MU, Weissleder R, Wick W, Heiland S, Vollmuth P, Bendszus M, Rodell CB, Breckwoldt MO, Platten M. T cell-independent eradication of experimental glioma by intravenous TLR7/8-agonist-loaded nanoparticles. Nat Commun 2023; 14:771. [PMID: 36774352 PMCID: PMC9922247 DOI: 10.1038/s41467-023-36321-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 01/24/2023] [Indexed: 02/13/2023] Open
Abstract
Glioblastoma, the most common and aggressive primary brain tumor type, is considered an immunologically "cold" tumor with sparse infiltration by adaptive immune cells. Immunosuppressive tumor-associated myeloid cells are drivers of tumor progression. Therefore, targeting and reprogramming intratumoral myeloid cells is an appealing therapeutic strategy. Here, we investigate a β-cyclodextrin nanoparticle (CDNP) formulation encapsulating the Toll-like receptor 7 and 8 (TLR7/8) agonist R848 (CDNP-R848) to reprogram myeloid cells in the glioma microenvironment. We show that intravenous monotherapy with CDNP-R848 induces regression of established syngeneic experimental glioma, resulting in increased survival rates compared with unloaded CDNP controls. Mechanistically, CDNP-R848 treatment reshapes the immunosuppressive tumor microenvironment and orchestrates tumor clearing by pro-inflammatory tumor-associated myeloid cells, independently of T cells and NK cells. Using serial magnetic resonance imaging, we identify a radiomic signature in response to CDNP-R848 treatment and ultrasmall superparamagnetic iron oxide (USPIO) imaging reveals that immunosuppressive macrophage recruitment is reduced by CDNP-R848. In conclusion, CDNP-R848 induces tumor regression in experimental glioma by targeting blood-borne macrophages without requiring adaptive immunity.
Collapse
Affiliation(s)
- Verena Turco
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany.,Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Kira Pfleiderer
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Jessica Hunger
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Natalie K Horvat
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, University Hospital, Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), Heidelberg University, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Kianush Karimian-Jazi
- Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Katharina Schregel
- Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Manuel Fischer
- Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Gianluca Brugnara
- Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Kristine Jähne
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Volker Sturm
- Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Yannik Streibel
- Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Duy Nguyen
- Junior Research Group Bioinformatics and Omics Data Analytics, DKFZ, Heidelberg, Germany
| | - Sandro Altamura
- Department of Pediatric Oncology, Hematology and Immunology, University Hospital, Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), Heidelberg University, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Dennis A Agardy
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Shreya S Soni
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Abdulrahman Alsasa
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Theresa Bunse
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany
| | - Matthias Schlesner
- Junior Research Group Bioinformatics and Omics Data Analytics, DKFZ, Heidelberg, Germany.,Biomedical Informatics, Data Mining and Data Analytics, Faculty of Applied Computer Science and Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Oncology, Hematology and Immunology, University Hospital, Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), Heidelberg University, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, DKTK within DKFZ, Heidelberg, Germany.,Department of Neurology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
| | - Sabine Heiland
- Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Philipp Vollmuth
- Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Martin Bendszus
- Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Christopher B Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Michael O Breckwoldt
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany. .,Neuroradiology Department, University Hospital Heidelberg, 69120, Heidelberg, Germany.
| | - Michael Platten
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany. .,Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim, Germany.
| |
Collapse
|
44
|
Jin GW, Choi G, Piao H, Rejinold NS, Asahina S, Choi SJ, Lee HJ, Choy JH. NOAEL cancer therapy: a tumor targetable docetaxel-inorganic polymer nanohybrid prevents drug-induced neutropenia. J Mater Chem B 2023; 11:565-575. [PMID: 36354057 DOI: 10.1039/d2tb02121h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
To date, cancer therapies largely consist of five pillars: surgery, radiation, chemotherapy, targeted therapy, and immunotherapy. Still, researchers are trying to innovate the current cancer therapies to pursue an ideal one without side effects. For developing such a therapy, we designed a chemically well-defined route to a PEG- and docetaxel (DTX)-conjugated inorganic polymer, polyphosphazene, named "polytaxel (PTX)" with a prolonged blood circulation time and tumor localization. Here, we conducted the proof-of-concept study of the ideal therapy in orthotopic and xenograft pancreatic cancer models. We found that the average tumor inhibition rates of PTX were similar to those of DTX without any DTX toxicity-related side effects, such as neutropenia and weight loss. In conclusion, PTX met the requirements of an ideal anticancer drug with high anticancer efficacy and 100% survival rate. PTX is expected to replace any existing anticancer therapies in clinical practice.
Collapse
Affiliation(s)
- Geun-Woo Jin
- R&D Center, CnPharm Co., Ltd, Seoul 03759, Republic of Korea
| | - Goeun Choi
- Intelligent Nanohybrid Materials Laboratory (INML), Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea. .,College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.,Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Huiyan Piao
- Intelligent Nanohybrid Materials Laboratory (INML), Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea.
| | - N Sanoj Rejinold
- Intelligent Nanohybrid Materials Laboratory (INML), Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea.
| | - Shunsuke Asahina
- JEOL Ltd. SM Business Unit, 3-1-2 Musashino, Akishima-shi, Tokyo 196-8558, Japan
| | - Soo-Jin Choi
- Division of Applied Food System, Major of Food Science & Technology, Seoul Women's University, Seoul 01797, Republic of Korea
| | - Hwa Jeong Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Jin-Ho Choy
- Intelligent Nanohybrid Materials Laboratory (INML), Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea. .,Division of Natural Sciences, The National Academy of Sciences, Seoul 06579, Republic of Korea.,Department of Pre-medical Course, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea.,International Research Frontier Initiative (IRFI), Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| |
Collapse
|
45
|
Donoso‐Meneses D, Figueroa‐Valdés AI, Georges N, Tobar HE, Alcayaga‐Miranda F. Turning adversity into opportunity: Small extracellular vesicles as nanocarriers for tumor-associated macrophages re-education. Bioeng Transl Med 2023; 8:e10349. [PMID: 36684102 PMCID: PMC9842057 DOI: 10.1002/btm2.10349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 01/25/2023] Open
Abstract
Currently, small extracellular vesicles (sEV) as a nanoscale drug delivery system, are undergoing biotechnological scaling and clinical validation. Nonetheless, preclinical pharmacokinetic studies revealed that sEV are predominantly uptaken by macrophages. Although this "sEV-macrophage" propensity represents a disadvantage in terms of sEV targeting and their bioavailability as nanocarriers, it also represents a strategic advantage for those therapies that involve macrophages. Such is the case of tumor-associated macrophages (TAMs), which can reprogram/repolarize their predominantly immunosuppressive and tumor-supportive phenotype toward an immunostimulatory and anti-tumor phenotype using sEV as nanocarriers of TAMs reprogramming molecules. In this design, sEV represents an advantageous delivery system, providing precision to the therapy by simultaneously matching their tropism to the therapeutic cell target. Here, we review the current knowledge of the role of TAMs in the tumoral microenvironment and the effect generated by the reprogramming of these phagocytic cells fate using sEV. Finally, we discuss how these vesicles can be engineered by different bioengineering techniques to improve their therapeutic cargo loading and preferential uptake by TAMs.
Collapse
Affiliation(s)
- Dario Donoso‐Meneses
- Laboratory of Nano‐Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of MedicineUniversidad de Los AndesSantiagoChile
- Consorcio RegeneroChilean Consortium for Regenerative MedicineSantiagoChile
- IMPACTCenter of Interventional Medicine for Precision and Advanced Cellular TherapySantiagoChile
| | - Aliosha I. Figueroa‐Valdés
- Laboratory of Nano‐Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of MedicineUniversidad de Los AndesSantiagoChile
- Consorcio RegeneroChilean Consortium for Regenerative MedicineSantiagoChile
- IMPACTCenter of Interventional Medicine for Precision and Advanced Cellular TherapySantiagoChile
| | - Nicolás Georges
- Laboratory of Nano‐Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of MedicineUniversidad de Los AndesSantiagoChile
- Consorcio RegeneroChilean Consortium for Regenerative MedicineSantiagoChile
- IMPACTCenter of Interventional Medicine for Precision and Advanced Cellular TherapySantiagoChile
| | - Hugo E. Tobar
- Laboratory of Nano‐Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of MedicineUniversidad de Los AndesSantiagoChile
- IMPACTCenter of Interventional Medicine for Precision and Advanced Cellular TherapySantiagoChile
| | - Francisca Alcayaga‐Miranda
- Laboratory of Nano‐Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of MedicineUniversidad de Los AndesSantiagoChile
- Consorcio RegeneroChilean Consortium for Regenerative MedicineSantiagoChile
- IMPACTCenter of Interventional Medicine for Precision and Advanced Cellular TherapySantiagoChile
- School of Medicine, Faculty of MedicineUniversidad de Los AndesSantiagoChile
- Cells for CellsSantiagoChile
| |
Collapse
|
46
|
Shin YB, Choi JY, Shin DH, Lee JW. Anticancer Evaluation of Methoxy Poly(Ethylene Glycol)- b-Poly(Caprolactone) Polymeric Micelles Encapsulating Fenbendazole and Rapamycin in Ovarian Cancer. Int J Nanomedicine 2023; 18:2209-2223. [PMID: 37152471 PMCID: PMC10162106 DOI: 10.2147/ijn.s394712] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/25/2023] [Indexed: 05/09/2023] Open
Abstract
Purpose We aimed to inhibit ovarian cancer (OC) development by interfering with microtubule polymerization and inhibiting mTOR signaling. To achieve this, previously developed micelles containing fenbendazole and rapamycin were applied. Methods Herein, we prepared micelles for drug delivery using fenbendazole and rapamycin at a 1:2 molar ratio and methoxy poly(ethylene glycol)-b-poly(caprolactone)(mPEG-b-PCL) via freeze-drying. We revealed their long-term storage capacity of up to 120 days. Furthermore, a cytotoxicity test was performed on the OC cell line HeyA8, and an orthotopic model was established for evaluating in vivo antitumor efficacy. Results Fenbendazole/rapamycin-loaded mPEG-b-PCL micelle (M-FR) had an average particle size of 37.2 ± 1.10 nm, a zeta potential of -0.07 ± 0.09 mV, and a polydispersity index of 0.20 ± 0.02. Additionally, the average encapsulation efficiency of fenbendazole was 75.7 ± 4.61% and that of rapamycin was 98.0 ± 1.97%. In the clonogenic assay, M-FR was 6.9 times more effective than that free fenbendazole/rapamycin. The in vitro drug release profile showed slower release in the combination formulation than in the single formulation. Conclusion There was no toxicity, and tumor growth was suppressed substantially by our formulation compared with that seen with the control. The findings of our study lay a foundation for using fenbendazole and rapamycin for OC treatment.
Collapse
Affiliation(s)
- Yu Been Shin
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Ju-Yeon Choi
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Dae Hwan Shin
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
- Correspondence: Dae Hwan Shin, College of Pharmacy, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, 28160, Republic of Korea, Tel +82 43 261 2820, Fax +82 43 268 2732, Email
| | - Jeong-Won Lee
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Jeong-Won Lee, Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea, Tel +82-2-3410-1382, Fax +82-2-3410-0630, Email
| |
Collapse
|
47
|
Singh N, Shi S, Goel S. Ultrasmall silica nanoparticles in translational biomedical research: Overview and outlook. Adv Drug Deliv Rev 2023; 192:114638. [PMID: 36462644 PMCID: PMC9812918 DOI: 10.1016/j.addr.2022.114638] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/06/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022]
Abstract
The exemplary progress of silica nanotechnology has attracted extensive attention across a range of biomedical applications such as diagnostics and imaging, drug delivery, and therapy of cancer and other diseases. Ultrasmall silica nanoparticles (USNs) have emerged as a particularly promising class demonstrating unique properties that are especially suitable for and have shown great promise in translational and clinical biomedical research. In this review, we discuss synthetic strategies that allow precise engineering of USNs with excellent control over size and surface chemistry, functionalization, and pharmacokinetic and toxicological profiles. We summarize the current state-of-the-art in the biomedical applications of USNs with a particular focus on select clinical studies. Finally, we illustrate long-standing challenges in the translation of inorganic nanotechnology, particularly in the context of ultrasmall nanomedicines, and provide our perspectives on potential solutions and future opportunities in accelerating the translation and widespread adoption of USN technology in biomedical research.
Collapse
Affiliation(s)
- Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112,Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112,Correspondence to ;
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112,Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112,Correspondence to ;
| |
Collapse
|
48
|
Chu Y, Luo Y, Su B, Li C, Guo Q, Zhang Y, Liu P, Chen H, Zhao Z, Zhou Z, Wang Y, Jiang C, Sun T. A neutrophil-biomimic platform for eradicating metastatic breast cancer stem-like cells by redox microenvironment modulation and hypoxia-triggered differentiation therapy. Acta Pharm Sin B 2023; 13:298-314. [PMID: 36815033 PMCID: PMC9939302 DOI: 10.1016/j.apsb.2022.05.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/10/2022] [Accepted: 05/15/2022] [Indexed: 11/18/2022] Open
Abstract
Metastasis accounts for 90% of breast cancer deaths, where the lethality could be attributed to the poor drug accumulation at the metastatic loci. The tolerance to chemotherapy induced by breast cancer stem cells (BCSCs) and their particular redox microenvironment further aggravate the therapeutic dilemma. To be specific, therapy-resistant BCSCs can differentiate into heterogeneous tumor cells constantly, and simultaneously dynamic maintenance of redox homeostasis promote tumor cells to retro-differentiate into stem-like state in response to cytotoxic chemotherapy. Herein, we develop a specifically-designed biomimic platform employing neutrophil membrane as shell to inherit a neutrophil-like tumor-targeting capability, and anchored chemotherapeutic and BCSCs-differentiating reagents with nitroimidazole (NI) to yield two hypoxia-responsive prodrugs, which could be encapsulated into a polymeric nitroimidazole core. The platform can actively target the lung metastasis sites of triple negative breast cancer (TNBC), and release the escorted drugs upon being triggered by the hypoxia microenvironment. During the responsiveness, the differentiating agent could promote transferring BCSCs into non-BCSCs, and simultaneously the nitroimidazole moieties conjugated on the polymer and prodrugs could modulate the tumor microenvironment by depleting nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) and amplifying intracellular oxidative stress to prevent tumor cells retro-differentiation into BCSCs. In combination, the BCSCs differentiation and tumor microenvironment modulation synergistically could enhance the chemotherapeutic cytotoxicity, and remarkably suppress tumor growth and lung metastasis. Hopefully, this work can provide a new insight in to comprehensively treat TNBC and lung metastasis using a versatile platform.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Chen Jiang
- Corresponding author. Tel./fax: +86 21 5198 0079.
| | - Tao Sun
- Corresponding author. Tel./fax: +86 21 5198 0079.
| |
Collapse
|
49
|
Famta P, Shah S, Jain N, Srinivasarao DA, Murthy A, Ahmed T, Vambhurkar G, Shahrukh S, Singh SB, Srivastava S. Albumin-hitchhiking: Fostering the pharmacokinetics and anticancer therapeutics. J Control Release 2023; 353:166-185. [PMID: 36423870 DOI: 10.1016/j.jconrel.2022.11.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
Nanotherapeutics demonstrate poor accumulation in the tumor microenvironment due to poor extravasation and penetration into the tumor. Therapeutics such as oligonucleotides, peptides and other biologicals suffer from low systemic half-life and rapid degradation. Albumin-hitchhiking has emerged as an effective strategy to enhance tumor-specific accumulation of various therapeutics. Hitchhiking on serum albumin (SA) have shown to improve biological half-life of various therapeutics including nanocarriers (NCs), biologics, oligonucleotides, vaccines, etc. In addition, passive and active accumulation of SA-riding therapeutics in the tumor, site-specific drug release, and SA-mediated endosomal escape have improved the potential of various anticancer modalities such as chemo-, immune-, vaccine, and gene therapies. In this review, we have discussed the advantages of employing SA-hitchhiking in anticancer therapies. In addition, vaccine strategies employing inherent lymph-nodes accumulating property of albumin have been discussed. We have presented a clinical overview of SA-hitchhiked formulations along with possible bottlenecks for improved clinical outcomes. We have also discussed the role of physiologically based pharmacokinetics (PBPK) modelling for efficient characterization of anti-cancer nanotherapeutics.
Collapse
Affiliation(s)
- Paras Famta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Naitik Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dadi A Srinivasarao
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Aditya Murthy
- Department of Biopharmaceutics and Bioequivalence, Dr. Reddy's Laboratories Ltd., Global Clinical Management Group, IPDO, Hyderabad, India
| | - Tausif Ahmed
- Department of Biopharmaceutics and Bioequivalence, Dr. Reddy's Laboratories Ltd., Global Clinical Management Group, IPDO, Hyderabad, India
| | - Ganesh Vambhurkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Syed Shahrukh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
50
|
Malinovskaya J, Salami R, Valikhov M, Vadekhina V, Semyonkin A, Semkina A, Abakumov M, Harel Y, Levy E, Levin T, Persky R, Chekhonin V, Lellouche JP, Melnikov P, Gelperina S. Supermagnetic Human Serum Albumin (HSA) Nanoparticles and PLGA-Based Doxorubicin Nanoformulation: A Duet for Selective Nanotherapy. Int J Mol Sci 2022; 24:ijms24010627. [PMID: 36614071 PMCID: PMC9820361 DOI: 10.3390/ijms24010627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Predicting the ability of nanoparticles (NP) to access the tumor is key to the success of chemotherapy using nanotherapeutics. In the present study, the ability of the dual NP-based theranostic system to accumulate in the tumor was evaluated in vivo using intravital microscopy (IVM) and MRI. The system consisted of model therapeutic doxorubicin-loaded poly(lactide-co-glycolide) NP (Dox-PLGA NP) and novel hybrid Ce3/4+-doped maghemite NP encapsulated within the HSA matrix (hMNP) as a supermagnetic MRI contrasting agent. Both NP types had similar sizes of ~100 nm and negative surface potentials. The level of the hMNP and PLGA NP co-distribution in the same regions of interest (ROI, ~2500 µm2) was assessed by IVM in mice bearing the 4T1-mScarlet murine mammary carcinoma at different intervals between the NP injections. In all cases, both NP types penetrated into the same tumoral/peritumoral regions by neutrophil-assisted extravasation through vascular micro- and macroleakages. The maximum tumor contrasting in MRI scans was obtained 5 h after hMNP injection/1 h after PLGA NP injection; the co-distribution level at this time reached 78%. Together with high contrasting properties of the hMNP, these data indicate that the hMNP and PLGA NPs are suitable theranostic companions. Thus, analysis of the co-distribution level appears to be a useful tool for evaluation of the dual nanoparticle theranostics, whereas assessment of the leakage areas helps to reveal the tumors potentially responsive to nanotherapeutics.
Collapse
Affiliation(s)
- Julia Malinovskaya
- Drug Delivery Systems Laboratory, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047 Moscow, Russia
| | - Rawan Salami
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Institute of Nanotechnology and Advanced Materials, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Marat Valikhov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ostrovityanova ul 1, 117997 Moscow, Russia
| | - Veronika Vadekhina
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034 Moscow, Russia
| | - Aleksey Semyonkin
- Drug Delivery Systems Laboratory, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047 Moscow, Russia
| | - Alevtina Semkina
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ostrovityanova ul 1, 117997 Moscow, Russia
| | - Maxim Abakumov
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ostrovityanova ul 1, 117997 Moscow, Russia
| | - Yifat Harel
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Institute of Nanotechnology and Advanced Materials, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Esthy Levy
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Institute of Nanotechnology and Advanced Materials, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Tzuriel Levin
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Institute of Nanotechnology and Advanced Materials, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Rachel Persky
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Vladimir Chekhonin
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034 Moscow, Russia
| | - Jean-Paul Lellouche
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Institute of Nanotechnology and Advanced Materials, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Pavel Melnikov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034 Moscow, Russia
| | - Svetlana Gelperina
- Drug Delivery Systems Laboratory, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047 Moscow, Russia
- Correspondence:
| |
Collapse
|