1
|
Bigaj-Józefowska MJ, Zalewski T, Załęski K, Coy E, Frankowski M, Mrówczyński R, Grześkowiak BF. Three musketeers of PDA-based MRI contrasting and therapy. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:321-333. [PMID: 38795050 DOI: 10.1080/21691401.2024.2356773] [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: 03/04/2024] [Accepted: 05/13/2024] [Indexed: 05/27/2024]
Abstract
Polydopamine (PDA) stands as a versatile material explored in cancer nanomedicine for its unique properties, offering opportunities for multifunctional drug delivery platforms. This study explores the potential of utilizing a one-pot synthesis to concurrently integrate Fe, Gd and Mn ions into porous PDA-based theranostic drug delivery platforms called Ferritis, Gadolinis and Manganis, respectively. Our investigation spans the morphology, magnetic properties, photothermal characteristics and cytotoxicity profiles of those potent nanoformulations. The obtained structures showcase a spherical morphology, robust magnetic response and promising photothermal behaviour. All of the presented nanoparticles (NPs) display pronounced paramagnetism, revealing contrasting potential for MRI imaging. Relaxivity values, a key determinant of contrast efficacy, demonstrated competitive or superior performance compared to established, used contrasting agents. These nanoformulations also exhibited robust photothermal properties under near infra-red irradiation, showcasing their possible application for photothermal therapy of cancer. Our findings provide insights into the potential of metal-doped PDA NPs for cancer theranostics.
Collapse
Affiliation(s)
| | - Tomasz Zalewski
- NanoBioMedical Centre, Adam Mickiewicz University in Poznań, Poznań, Poland
| | - Karol Załęski
- NanoBioMedical Centre, Adam Mickiewicz University in Poznań, Poznań, Poland
| | - Emerson Coy
- NanoBioMedical Centre, Adam Mickiewicz University in Poznań, Poznań, Poland
| | - Marcin Frankowski
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, Poznań, Poland
| | - Radosław Mrówczyński
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, Poznań, Poland
- Centre for Advanced Technologies, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | |
Collapse
|
2
|
Liu H, Xiong H, Li C, Xu M, Yun Y, Ruan Y, Tang L, Zhang T, Su D, Sun X. 131I Induced In Vivo Proteolysis by Photoswitchable azoPROTAC Reinforces Internal Radiotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310865. [PMID: 38678537 DOI: 10.1002/smll.202310865] [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: 11/24/2023] [Revised: 03/18/2024] [Indexed: 05/01/2024]
Abstract
Photopharmacology, incorporating photoswitches such as azobenezes into drugs, is an emerging therapeutic method to realize spatiotemporal control of pharmacological activity by light. However, most photoswitchable molecules are triggered by UV light with limited tissue penetration, which greatly restricts the in vivo application. Here, this study proves that 131I can trigger the trans-cis photoisomerization of a reported azobenezen incorporating PROTACs (azoPROTAC). With the presence of 50 µCi mL-1 131I, the azoPROTAC can effectively down-regulate BRD4 and c-Myc levels in 4T1 cells at a similar level as it does under light irradiation (405 nm, 60 mW cm-2). What's more, the degradation of BRD4 can further benefit the 131I-based radiotherapy. The in vivo experiment proves that intratumoral co-adminstration of 131I (300 µCi) and azoPROTC (25 mg kg-1) via hydrogel not only successfully induce protein degradation in 4T1 tumor bearing-mice but also efficiently inhibit tumor growth with enhanced radiotherapeutic effect and anti-tumor immunological effect. This is the first time that a radioisotope is successfully used as a trigger in photopharmacology in a mouse model. It believes that this study will benefit photopharmacology in deep tissue.
Collapse
Affiliation(s)
- Huihui Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, 621000, China
| | - Hehua Xiong
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Changjun Li
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Mengxia Xu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuyang Yun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yiling Ruan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lijun Tang
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Tao Zhang
- Department of Radiopharmaceuticals, Nuclear Medicine Clinical Translation Center, Nanjing Medical University, Nanjing, 211166, China
| | - Dan Su
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Department of Clinical Medicine, Hangzhou Medical College, Hangzhou, 310053, China
| | - Xiaolian Sun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
3
|
Hübinger L, Wetzig K, Runge R, Hartmann H, Tillner F, Tietze K, Pretze M, Kästner D, Freudenberg R, Brogsitter C, Kotzerke J. Investigation of Photodynamic Therapy Promoted by Cherenkov Light Activated Photosensitizers-New Aspects and Revelations. Pharmaceutics 2024; 16:534. [PMID: 38675195 PMCID: PMC11054706 DOI: 10.3390/pharmaceutics16040534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
This work investigates the proposed enhanced efficacy of photodynamic therapy (PDT) by activating photosensitizers (PSs) with Cherenkov light (CL). The approaches of Yoon et al. to test the effect of CL with external radiation were taken up and refined. The results were used to transfer the applied scheme from external radiation therapy to radionuclide therapy in nuclear medicine. Here, the CL for the activation of the PSs (psoralen and trioxsalen) is generated by the ionizing radiation from rhenium-188 (a high-energy beta-emitter, Re-188). In vitro cell survival studies were performed on FaDu, B16 and 4T1 cells. A characterization of the PSs (absorbance measurement and gel electrophoresis) and the CL produced by Re-188 (luminescence measurement) was performed as well as a comparison of clonogenic assays with and without PSs. The methods of Yoon et al. were reproduced with a beam line at our facility to validate their results. In our studies with different concentrations of PS and considering the negative controls without PS, the statements of Yoon et al. regarding the positive effect of CL could not be confirmed. There are slight differences in survival fractions, but they are not significant when considering the differences in the controls. Gel electrophoresis showed a dominance of trioxsalen over psoralen in conclusion of single and double strand breaks in plasmid DNA, suggesting a superiority of trioxsalen as a PS (when irradiated with UVA). In addition, absorption measurements showed that these PSs do not need to be shielded from ambient light during the experiment. An observational test setup for a PDT nuclear medicine approach was found. The CL spectrum of Re-188 was measured. Fluctuating inconclusive results from clonogenic assays were found.
Collapse
Affiliation(s)
- Lisa Hübinger
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Kerstin Wetzig
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Roswitha Runge
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Holger Hartmann
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Falk Tillner
- Department of Radiation Therapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden—Rossendorf, Institute of Radiooncology—OncoRay, 01328 Dresden, Germany
| | - Katja Tietze
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Marc Pretze
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - David Kästner
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Robert Freudenberg
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Claudia Brogsitter
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Jörg Kotzerke
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
4
|
Egbulefu C, Black K, Su X, Karmakar P, Habimana-Griffin L, Sudlow G, Prior J, Chukwu E, Zheleznyak A, Xu B, Xu Y, Esser A, Mixdorf M, Moss E, Manion B, Reed N, Gubin M, Lin CY, Schreiber R, Weilbaecher K, Achilefu S. Induction of complementary immunogenic necroptosis and apoptosis cell death pathways inhibits cancer metastasis and relapse. RESEARCH SQUARE 2024:rs.3.rs-3992212. [PMID: 38558990 PMCID: PMC10980095 DOI: 10.21203/rs.3.rs-3992212/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Interactions of light-sensitive drugs and materials with Cerenkov radiation-emitting radiopharmaceuticals generate cytotoxic reactive oxygen species (ROS) to inhibit localized and disseminated cancer progression, but the cell death mechanisms underlying this radionuclide stimulated dynamic therapy (RaST) remain elusive. Using ROS-regenerative nanophotosensitizers coated with a tumor-targeting transferrin-titanocene complex (TiO2-TC-Tf) and radiolabeled 2-fluorodeoxyglucose (18FDG), we found that adherent dying cells maintained metabolic activity with increased membrane permeabilization. Mechanistic assessment of these cells revealed that RaST activated the expression of RIPK-1 and RIPK-3, which mediate necroptosis cell death. Subsequent recruitment of the nuclear factors kappa B and the executioner mixed lineage kinase domain-like pseudo kinase (MLKL) triggered plasma membrane permeabilization and pore formation, respectively, followed by the release of cytokines and immunogenic damage-associated molecular patterns (DAMPs). In immune-deficient breast cancer models with adequate stroma and growth factors that recapitulate the human tumor microenvironment, RaST failed to inhibit tumor progression and the ensuing lung metastasis. A similar aggressive tumor model in immunocompetent mice responded to RaST, achieving a remarkable partial response (PR) and complete response (CR) with no evidence of lung metastasis, suggesting active immune system engagement. RaST recruited antitumor CD11b+, CD11c+, and CD8b+ effector immune cells after initiating dual immunogenic apoptosis and necroptosis cell death pathways in responding tumors in vivo. Over time, cancer cells upregulated the expression of negative immune regulating cytokine (TGF-β) and soluble immune checkpoints (sICP) to challenge RaST effect in the CR mice. Using a signal-amplifying cancer-imaging agent, LS301, we identified latent minimal residual disseminated tumors in the lymph nodes (LNs) of the CR group. Despite increased protumor immunogens in the CR mice, RaST prevented cancer relapse and metastasis through dynamic redistribution of ROS-regenerative TiO2 from bones at the early treatment stage to the spleen and LNs, maintaining active immunity against cancer progression and migration. This study reveals the immune-mechanistic underpinnings of RaST-mediated antitumor immune response and highlights immunogenic reprogramming of tumors in response to RaST. Overcoming apoptosis resistance through complementary necroptosis activation paves the way for strategic drug combinations to improve cancer treatment.
Collapse
Affiliation(s)
- Christopher Egbulefu
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75235-9397, USA
| | - Kvar Black
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Xinming Su
- Department of Medicine, Washington University in St. Louis, MO 63110, USA
| | - Partha Karmakar
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | | | - Gail Sudlow
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Julie Prior
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Ezeugo Chukwu
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Alex Zheleznyak
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Baogang Xu
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Yalin Xu
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Alison Esser
- Department of Medicine, Washington University in St. Louis, MO 63110, USA
| | - Matthew Mixdorf
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Evan Moss
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Brad Manion
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Nathan Reed
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
| | - Matthew Gubin
- Department of Pathology and Immunology, Washington University in St. Louis, MO 63110, USA
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington University in St. Louis, MO 63110, USA
| | - Robert Schreiber
- Department of Pathology and Immunology, Washington University in St. Louis, MO 63110, USA
| | | | - Samuel Achilefu
- Department of Radiology, Washington University in St. Louis, MO 63110, USA
- Department of Medicine, Washington University in St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63110, USA
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75235-9397, USA
| |
Collapse
|
5
|
Gendron LN, Sheveland CG, Gunn JR, Pogue BW, Shell TA, Shell JR. Radiation-Activated Cobalamin-Kinase Inhibitors for Treatment of Pancreatic Ductal Adenocarcinoma. Mol Pharm 2024; 21:137-142. [PMID: 37989273 PMCID: PMC11228961 DOI: 10.1021/acs.molpharmaceut.3c00667] [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] [Indexed: 11/23/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most dismal diagnoses that a patient can receive. PDAC is extremely difficult to treat, as drug delivery is challenging in part due to the lack of vascularization, high stromal content, and high collagen content of these tumors. We have previously demonstrated that attaching drugs to the cobalamin scaffold provides selectivity for tumors over benign cells due to a high vitamin demand in these rapidly growing cells and an overexpression of transcobalamin receptors in a variety of cancer types. Importantly, we have shown the ability to deliver cobalamin derivatives to orthotopic pancreas tumors. Tyrosine kinase inhibitors have shown promise in treating PDAC as well as other cancer types. However, some of these inhibitors suffer from drug resistance, and as such, their success has been diminished. With this in mind, we synthesized the tyrosine kinase inhibitors erlotinib (EGFR) and dasatinib (Src) that are attached to this cobalamin platform. Both of these cobalamin-drug conjugates cause visible light-induced apoptosis, and the cobalamin-erlotinib conjugate (2) causes X-ray-induced apoptosis in MIA PaCa-2 cells. Both visible light and X-rays provide spatial control of drug release; however, utilizing X-ray irradiation offers the advantage of deeper tissue penetration. Therefore, we explored the utilization of 2 as a synergistic therapy with radiation in athymic nude mice implanted with MIA PaCa-2 tumors. We discovered that the addition of 2 caused an enhanced reduction in tumor margins in comparison with radiation therapy alone. In addition, treatment with 2 in the absence of radiation caused no significant reduction in tumor size in comparison with the controls. The cobalamin technology presented here allows for the spatial release of drugs in conjunction with external beam radiation therapy, potentially allowing for more effective treatment of deep-seated tumors with less systemic side effects.
Collapse
Affiliation(s)
- Liberty N Gendron
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Colter G Sheveland
- Department of Chemistry and Biochemistry, Norwich University, Northfield, Vermont 05663, United States
| | - Jason R Gunn
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Brian W Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Thomas A Shell
- Department of Chemistry and Physics, Lincoln Memorial University, Harrogate, Tennessee 37752, United States
| | - Jennifer R Shell
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
- Eos Pharmaceuticals LLC, Tazewell, Tennessee 37879, United States
| |
Collapse
|
6
|
An Y, Xu D, Wen X, Chen C, Liu G, Lu Z. Internal Light Sources-Mediated Photodynamic Therapy Nanoplatforms: Hope for the Resolution of the Traditional Penetration Problem. Adv Healthc Mater 2024; 13:e2301326. [PMID: 37413664 DOI: 10.1002/adhm.202301326] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023]
Abstract
Photodynamic therapy (PDT) is an alternative cancer treatment technique with a noninvasive nature, high selectivity, and minimal adverse effects. The indispensable light source used in PDT is a critical factor in determining the energy conversion of photosensitizers (PSs). Traditional light sources are primarily concentrated in the visible light region, severely limiting their penetration depth and making them prone to scattering and absorption when applied to biological tissues. For that reason, its efficacy in treating deep-seated lesions is often inadequate. Self-exciting PDT, also known as auto-PDT (APDT), is an attractive option for circumventing the limited penetration depth of traditional PDT and has acquired significant attention. APDT employs depth-independent internal light sources to excite PSs through resonance or radiative energy transfer. APDT has considerable potential for treating deep-tissue malignancies. To facilitate many researchers' comprehension of the latest research progress in this field and inspire the emergence of more novel research results. This review introduces internal light generation mechanisms and characteristics and provides an overview of current research progress based on the recently reported APDT nanoplatforms. The current challenges and possible solutions of APDT nanoplatforms are also presented and provide insights for future research in the final section of this article.
Collapse
Affiliation(s)
- Yibo An
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Dazhuang Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Xiaofei Wen
- Department of Interventional Radiology, The First Affilited Hospital of Xiamen University, Xiamen, 361000, China
| | - Chuan Chen
- Department of Pharmacy, Xiamen Medical College, Xiamen, 361023, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Zhixiang Lu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
7
|
Adhalrao SB, Jadhav KR, Patil PL, Kadam VJ, Nirmal MK. Engineering Platelet Membrane Imitating Nanoparticles for Targeted Therapeutic Delivery. Curr Pharm Biotechnol 2024; 25:1230-1244. [PMID: 37539932 DOI: 10.2174/1389201024666230804140926] [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: 03/17/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 08/05/2023]
Abstract
Platelet Membrane Imitating Nanoparticles (PMINs) is a novel drug delivery system that imitates the structure and functionality of platelet membranes. PMINs imitate surface markers of platelets to target specific cells and transport therapeutic cargo. PMINs are engineered by incorporating the drug into the platelet membrane and encapsulating it in a nanoparticle scaffold. This allows PMINs to circulate in the bloodstream and bind to target cells with high specificity, reducing off-target effects and improving therapeutic efficacy. The engineering of PMINs entails several stages, including the separation and purification of platelet membranes, the integration of therapeutic cargo into the membrane, and the encapsulation of the membrane in a nanoparticle scaffold. In addition to being involved in a few pathological conditions including cancer, atherosclerosis, and rheumatoid arthritis, platelets are crucial to the body's physiological processes. This study includes the preparation and characterization of platelet membrane-like nanoparticles and focuses on their most recent advancements in targeted therapy for conditions, including cancer, immunological disorders, atherosclerosis, phototherapy, etc. PMINs are a potential drug delivery system that combines the advantages of platelet membranes with nanoparticles. The capacity to create PMMNs with particular therapeutic cargo and surface markers provides new possibilities for targeted medication administration and might completely change the way that medicine is practiced. Despite the need for more studies to optimize the engineering process and evaluate the effectiveness and safety of PMINs in clinical trials, this technology has a lot of potential.
Collapse
Affiliation(s)
- Shradha B Adhalrao
- Department of Pharmaceutics, Bharati Vidyapeeth College of Pharmacy, Sector 8 CBD Belapur, Navi Mumbai - 400614, Maharashtra, India
| | - Kisan R Jadhav
- Department of Pharmaceutics, Bharati Vidyapeeth College of Pharmacy, Sector 8 CBD Belapur, Navi Mumbai - 400614, Maharashtra, India
| | - Prashant L Patil
- Department of Pharmaceutics, Bharati Vidyapeeth College of Pharmacy, Sector 8 CBD Belapur, Navi Mumbai - 400614, Maharashtra, India
| | - Vilasrao J Kadam
- Department of Pharmaceutics, Bharati Vidyapeeth College of Pharmacy, Sector 8 CBD Belapur, Navi Mumbai - 400614, Maharashtra, India
| | - M Kasekar Nirmal
- Department of Pharmaceutics, Bharati Vidyapeeth College of Pharmacy, Sector 8 CBD Belapur, Navi Mumbai - 400614, Maharashtra, India
| |
Collapse
|
8
|
Gawne PJ, Ferreira M, Papaluca M, Grimm J, Decuzzi P. New Opportunities and Old Challenges in the Clinical translation of Nanotheranostics. NATURE REVIEWS. MATERIALS 2023; 8:783-798. [PMID: 39022623 PMCID: PMC11251001 DOI: 10.1038/s41578-023-00581-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/27/2023] [Indexed: 07/20/2024]
Abstract
Nanoparticle-based systems imbued with both diagnostic and therapeutic functions, known as nanotheranostics, have enabled remarkable progress in guiding focal therapy, inducing active responses to endogenous and exogenous biophysical stimuli, and stratifying patients for optimal treatment. However, although in recent years more nanotechnological platforms and techniques have been implemented in the clinic, several important challenges remain that are specific to nanotheranostics. In this Review, we first discuss some of the many ways of 'constructing' nanotheranostics, focusing on the different imaging modalities and therapeutic strategies. We then outline nanotheranostics that are currently used in humans at different stages of clinical development, identifying specific advantages and opportunities. Finally, we define critical steps along the winding road of preclinical and clinical development and suggest actions to overcome technical, manufacturing, regulatory and economical challenges for the safe and effective clinical translation of nanotheranostics.
Collapse
Affiliation(s)
- Peter J. Gawne
- UCL Cancer Institute, University College London, London, UK
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary, University of London, London, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Miguel Ferreira
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Marisa Papaluca
- School of Public Health, Imperial College of London, South Kensington CampusLondon, UK
| | - Jan Grimm
- Molecular Pharmacology Program and Department of Radiology, Memorial Sloan-Kettering Cancer, Center, New York, NY, USA
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via, Morego 30, 16163, Genoa, IT
| |
Collapse
|
9
|
Lioret V, Bellaye PS, Bernhard Y, Moreau M, Guillemin M, Drouet C, Collin B, Decréau RA. Cherenkov Radiation induced photodynamic therapy - repurposing older photosensitizers, and radionuclides. Photodiagnosis Photodyn Ther 2023; 44:103816. [PMID: 37783257 DOI: 10.1016/j.pdpdt.2023.103816] [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: 03/21/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
CONTEXT Old-generation photosensitizers are minimally used in current photodynamic therapy (PDT) because they absorb in the UV/blue/green region of the spectrum where biological tissues are generally highly absorbing. The UV/blue light of Cherenkov Radiation (CR) from nuclear disintegration of beta-emitter radionuclides shows promise as an internal light source to activate these photosensitizers within tissue. Outline of the study: 1) radionuclide choice and Cherenkov Radiation, 2) Photosensitizer choice, synthesis and radiolabeling, 3) CR-induced fluorescence, 4) Verification of ROS formation, 5) CR-induced PDT with either free eosine and free CR emitter, or with radiolabelled eosin. RESULTS Cherenkov Radiation Energy Transfer (CRET) from therapeutic radionuclides (90Y) and PET imaging radionuclides (18F, 68Ga) to eosin was shown by spectrofluorimetry and in vitro, and was shown to result in a PDT process. The feasibility of CR-induced PDT (CR-PDT) was demonstrated in vitro on B16F10 murine melanoma cells mixing free eosin (λabs = 524 nm, ΦΔ 0.67) with free CR-emitter [18F]-FDG under their respective intrinsic toxicity levels (0.5 mM/8 MBq) and by trapping singlet oxygen with diphenylisobenzofuran (DPBF). An eosin-DOTAGA-chelate conjugate 1 was synthesized and radiometallated with CR-emitter [68Ga] allowed to reach 25 % cell toxicity at 0.125 mM/2 MBq, i.e. below the toxicity threshold of each component measured on controls. Incubation time was carefully examined, especially for CR emitters, in light of its toxicity, and its CR-emitting yield expected to be 3 times as much for 68Ga than 18F (considering their β particle energy) per radionuclide decay, while its half-life is about twice as small. PERSPECTIVE This study showed that in complete darkness, as it is at depth in tissues, PDT could proceed relying on CR emission from radionuclides only. Interestingly, this study also repurposed PET imaging radionuclides, such as 68Ga, to trigger a therapeutic event (PDT), albeit in a modest extent. Moreover, although it remains modest, such a PDT approach may be used to achieve additional tumoricidal effect to RIT treatment, where radionuclides, such as 90Y, are strong CR emitters, i.e. very potent light source for photosensitizer activation.
Collapse
Affiliation(s)
- Vivian Lioret
- ICMUB Institute (Chemistry Department) Sciences Mirande, Université de Bourgogne Franche Comté, 9 Avenue Alain Savary, Dijon 21078, France
| | | | - Yann Bernhard
- ICMUB Institute (Chemistry Department) Sciences Mirande, Université de Bourgogne Franche Comté, 9 Avenue Alain Savary, Dijon 21078, France
| | - Mathieu Moreau
- ICMUB Institute (Chemistry Department) Sciences Mirande, Université de Bourgogne Franche Comté, 9 Avenue Alain Savary, Dijon 21078, France
| | - Mélanie Guillemin
- Centre George François Leclerc, 1 rue du Professeur Marion, Dijon 21079, France
| | - Camille Drouet
- Centre George François Leclerc, 1 rue du Professeur Marion, Dijon 21079, France
| | - Bertrand Collin
- ICMUB Institute (Chemistry Department) Sciences Mirande, Université de Bourgogne Franche Comté, 9 Avenue Alain Savary, Dijon 21078, France; Centre George François Leclerc, 1 rue du Professeur Marion, Dijon 21079, France
| | - Richard A Decréau
- ICMUB Institute (Chemistry Department) Sciences Mirande, Université de Bourgogne Franche Comté, 9 Avenue Alain Savary, Dijon 21078, France.
| |
Collapse
|
10
|
Liu H, Wang Q, Guo J, Feng K, Ruan Y, Zhang Z, Ji X, Wang J, Zhang T, Sun X. Prodrug-based strategy with a two-in-one liposome for Cerenkov-induced photodynamic therapy and chemotherapy. J Control Release 2023; 364:206-215. [PMID: 37884209 DOI: 10.1016/j.jconrel.2023.10.036] [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: 07/08/2023] [Revised: 09/14/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Cerenkov radiation induced photodynamic therapy (CR-PDT) can tackle the tissue penetration limitation of traditional PDT. However, co-delivery of radionuclides and photosensitizer may cause continuous phototoxicity in normal tissues during the circulation. 5-aminolevulinic acid (ALA) which can intracellularly transform into photosensitive protoporphyrin IX (PpIX) is a cancer-selective photosensitizer with negligible side effect. However, the hydrophilic nature of ALA and the further conversion of PpIX to photoinactive Heme severely hinder the therapeutic benefits of ALA-based PDT. Herein, we developed an 89Zr-labeled, pH responsive ALA and artemisinin (ART) co-loaded liposome (89Zr-ALA-Liposome-ART) for highly selective cancer therapy. 89Zr can serve as the internal excitation source to self-activate PpIX for CR-PDT, and the photoinactive Heme can activate the chemotherapeutic effect of ART. The 89Zr-ALA-Liposome-ART exhibited excellent tumor inhibition capability in subcutaneous 4T1-tumor-bearing Balb/c mice via CR-PDT and chemotherapy. Combined with anti-PD-L1, the 89Zr-ALA-Liposome-ART elicited strong antitumor immunity to against tumor recurrence.
Collapse
Affiliation(s)
- Huihui Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Qing Wang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jingru Guo
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Kai Feng
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yiling Ruan
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Zhihao Zhang
- Department of Radiopharmaceuticals, Nuclear Medicine Clinical Translation Center, School of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China
| | - Xin Ji
- Department of Radiopharmaceuticals, Nuclear Medicine Clinical Translation Center, School of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China
| | - Jigang Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Tao Zhang
- Department of Radiopharmaceuticals, Nuclear Medicine Clinical Translation Center, School of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China.
| | - Xiaolian Sun
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
11
|
Aung W, Tsuji AB, Rikiyama K, Nishikido F, Obara S, Higashi T. Imaging assessment of photosensitizer emission induced by radionuclide-derived Cherenkov radiation using charge-coupled device optical imaging and long-pass filters. World J Radiol 2023; 15:315-323. [PMID: 38058603 PMCID: PMC10696188 DOI: 10.4329/wjr.v15.i11.315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/26/2023] [Accepted: 11/17/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Radionuclides produce Cherenkov radiation (CR), which can potentially activate photosensitizers (PSs) in phototherapy. Several groups have studied Cherenkov energy transfer to PSs using optical imaging; however, cost-effectively identifying whether PSs are excited by radionuclide-derived CR and detecting fluorescence emission from excited PSs remain a challenge. Many laboratories face the need for expensive dedicated equipment. AIM To cost-effectively confirm whether PSs are excited by radionuclide-derived CR and distinguish fluorescence emission from excited PSs. METHODS The absorbance and fluorescence spectra of PSs were measured using a microplate reader and fluorescence spectrometer to examine the photo-physical properties of PSs. To mitigate the need for expensive dedicated equipment and achieve the aim of the study, we developed a method that utilizes a charge-coupled device optical imaging system and appropriate long-pass filters of different wavelengths (manual sequential application of long-pass filters of 515, 580, 645, 700, 750, and 800 nm). Tetrakis (4-carboxyphenyl) porphyrin (TCPP) was utilized as a model PS. Different doses of copper-64 (64CuCl2) (4, 2, and 1 mCi) were used as CR-producing radionuclides. Imaging and data acquisition were performed 0.5 h after sample preparation. Differential image analysis was conducted by using ImageJ software (National Institutes of Health) to visually evaluate TCPP fluorescence. RESULTS The maximum absorbance of TCPP was at 390-430 nm, and the emission peak was at 670 nm. The CR and CR-induced TCPP emissions were observed using the optical imaging system and the high-transmittance long-pass filters described above. The emission spectra of TCPP with a peak in the 645-700 nm window were obtained by calculation and subtraction based on the serial signal intensity (total flux) difference between 64CuCl2 + TCPP and 64CuCl2. Moreover, the differential fluorescence images of TCPP were obtained by subtracting the 64CuCl2 image from the 64CuCl2 + TCPP image. The experimental results considering different 64CuCl2 doses showed a dose-dependent trend. These results demonstrate that a bioluminescence imaging device coupled with different long-pass filters and subtraction image processing can confirm the emission spectra and differential fluorescence images of CR-induced TCPP. CONCLUSION This simple method identifies the PS fluorescence emission generated by radionuclide-derived CR and can contribute to accelerating the development of Cherenkov energy transfer imaging and the discovery of new PSs.
Collapse
Affiliation(s)
- Winn Aung
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Atsushi B Tsuji
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Kazuaki Rikiyama
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Fumihiko Nishikido
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Satoshi Obara
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Tatsuya Higashi
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| |
Collapse
|
12
|
Cui X, Li X, Peng C, Qiu Y, Shi Y, Liu Y, Fei JF. Beyond External Light: On-Spot Light Generation or Light Delivery for Highly Penetrated Photodynamic Therapy. ACS NANO 2023; 17:20776-20803. [PMID: 37874930 DOI: 10.1021/acsnano.3c05619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
External light sources, such as lasers, light emitting diodes (LEDs) and lamps, are widely applied in photodynamic therapy (PDT); however, their use is severely limited by the nature of shallow tissue penetration depth. The recent exploration of light delivery or local generation on tumor sites has attracted much attention, owing to the fact that these systems are significantly endowed with high tissue penetration. In this review, we briefly introduced the principle of "on-spot light generation or delivery systems" in PDT. These systems are divided into different categories: (1) implantable luminescence, (2) mechanoluminescence, (3) electrochemiluminescence, (4) Cerenkov luminescence, (5) chemiluminescence, and (6) bioluminescence. Finally, their applications, advantages, and disadvantages in PDT will be appropriately summarized and further discussed in detail. We believe that this review will provide general guidance for the further design of light generation or delivery systems and clinical studies for PDT-mediated cancer treatments with unparalleled merits.
Collapse
Affiliation(s)
- Xiao Cui
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, People's Republic of China
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Xiang Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, People's Republic of China
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Cheng Peng
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, People's Republic of China
| | - Yuanhui Qiu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, People's Republic of China
| | - Yu Shi
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, People's Republic of China
| | - Yanmei Liu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, People's Republic of China
| | - Ji-Feng Fei
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, People's Republic of China
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, People's Republic of China
| |
Collapse
|
13
|
Gong Z, Chen J, Chen R, Zhu X, Wang C, Zhang X, Hu H, Yang Y, Zhang B, Chen H, Kaminer I, Lin X. Interfacial Cherenkov radiation from ultralow-energy electrons. Proc Natl Acad Sci U S A 2023; 120:e2306601120. [PMID: 37695899 PMCID: PMC10515145 DOI: 10.1073/pnas.2306601120] [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: 04/23/2023] [Accepted: 08/11/2023] [Indexed: 09/13/2023] Open
Abstract
Cherenkov radiation occurs only when a charged particle moves with a velocity exceeding the phase velocity of light in that matter. This radiation mechanism creates directional light emission at a wide range of frequencies and could facilitate the development of on-chip light sources except for the hard-to-satisfy requirement for high-energy particles. Creating Cherenkov radiation from low-energy electrons that has no momentum mismatch with light in free space is still a long-standing challenge. Here, we report a mechanism to overcome this challenge by exploiting a combined effect of interfacial Cherenkov radiation and umklapp scattering, namely the constructive interference of light emission from sequential particle-interface interactions with specially designed (umklapp) momentum-shifts. We find that this combined effect is able to create the interfacial Cherenkov radiation from ultralow-energy electrons, with kinetic energies down to the electron-volt scale. Due to the umklapp scattering for the excited high-momentum Bloch modes, the resulting interfacial Cherenkov radiation is uniquely featured with spatially separated apexes for its wave cone and group cone.
Collapse
Affiliation(s)
- Zheng Gong
- Interdisciplinary Center for Quantum Information, State Key Laboratory of Extreme Photonics and Instrumentation, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou310027, China
- International Joint Innovation Center, The Electromagnetics Academy at Zhejiang University, Zhejiang University, Haining314400, China
| | - Jialin Chen
- Interdisciplinary Center for Quantum Information, State Key Laboratory of Extreme Photonics and Instrumentation, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou310027, China
- International Joint Innovation Center, The Electromagnetics Academy at Zhejiang University, Zhejiang University, Haining314400, China
- Department of Electrical and Computer Engineering, Technion-Israel Institute of Technology, Haifa32000, Israel
| | - Ruoxi Chen
- Interdisciplinary Center for Quantum Information, State Key Laboratory of Extreme Photonics and Instrumentation, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou310027, China
- International Joint Innovation Center, The Electromagnetics Academy at Zhejiang University, Zhejiang University, Haining314400, China
| | - Xingjian Zhu
- School of Physics, Zhejiang University, Hangzhou310027, China
| | - Chan Wang
- Interdisciplinary Center for Quantum Information, State Key Laboratory of Extreme Photonics and Instrumentation, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou310027, China
- Key Laboratory of Advanced Micro/Nano Electronic Devices & Smart Systems of Zhejiang, Jinhua Institute of Zhejiang University, Zhejiang University, Jinhua321099, China
| | - Xinyan Zhang
- Interdisciplinary Center for Quantum Information, State Key Laboratory of Extreme Photonics and Instrumentation, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou310027, China
- International Joint Innovation Center, The Electromagnetics Academy at Zhejiang University, Zhejiang University, Haining314400, China
| | - Hao Hu
- College of Electronic and Information Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing211106, China
| | - Yi Yang
- Department of Physics, University of Hong Kong, Hong Kong999077, China
| | - Baile Zhang
- Division of Physics and Applied Physics, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore637371, Singapore
- Centre for Disruptive Photonic Technologies, Nanyang Technological University, Singapore637371, Singapore
| | - Hongsheng Chen
- Interdisciplinary Center for Quantum Information, State Key Laboratory of Extreme Photonics and Instrumentation, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou310027, China
- International Joint Innovation Center, The Electromagnetics Academy at Zhejiang University, Zhejiang University, Haining314400, China
- Key Laboratory of Advanced Micro/Nano Electronic Devices & Smart Systems of Zhejiang, Jinhua Institute of Zhejiang University, Zhejiang University, Jinhua321099, China
- Shaoxing Institute of Zhejiang University, Zhejiang University, Shaoxing312000, China
| | - Ido Kaminer
- Department of Electrical and Computer Engineering, Technion-Israel Institute of Technology, Haifa32000, Israel
| | - Xiao Lin
- Interdisciplinary Center for Quantum Information, State Key Laboratory of Extreme Photonics and Instrumentation, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou310027, China
- International Joint Innovation Center, The Electromagnetics Academy at Zhejiang University, Zhejiang University, Haining314400, China
| |
Collapse
|
14
|
Rosenkrans ZT, Hsu JC, Aluicio-Sarduy E, Barnhart TE, Engle JW, Cai W. Amplification of Cerenkov luminescence using semiconducting polymers for cancer theranostics. ADVANCED FUNCTIONAL MATERIALS 2023; 33:2302777. [PMID: 37942189 PMCID: PMC10629852 DOI: 10.1002/adfm.202302777] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Indexed: 11/10/2023]
Abstract
The therapeutic efficacy of photodynamic therapy is limited by the ability of light to penetrate tissues. Due to this limitation, Cerenkov luminescence (CL) from radionuclides has recently been proposed as an alternative light source in a strategy referred to as Cerenkov radiation induced therapy (CRIT). Semiconducting polymer nanoparticles (SPNs) have ideal optical properties, such as large absorption cross-sections and broad absorbance, which can be utilized to harness the relatively weak CL produced by radionuclides. SPNs can be doped with photosensitizers and have nearly 100% energy transfer efficiency by multiple energy transfer mechanisms. Herein, we investigated an optimized photosensitizer doped SPN as a nanosystem to harness and amplify CL for cancer theranostics. We found that semiconducting polymers significantly amplified CL energy transfer efficiency. Bimodal PET and optical imaging studies showed high tumor uptake and retention of the optimized SPNs when administered intravenously or intratumorally. Lastly, we found that photosensitizer doped SPNs have excellent potential as a cancer theranostics nanosystem in an in vivo tumor therapy study. Our study shows that SPNs are ideally suited to harness and amplify CL for cancer theranostics, which may provide a significant advancement for CRIT that are unabated by tissue penetration limits.
Collapse
Affiliation(s)
- Zachary T Rosenkrans
- University of Wisconsin-Madison, Department of Pharmaceutical Sciences, 600 Highland Ave., K6/562, Madison, WI 53792, USA
| | - Jessica C Hsu
- University of Wisconsin-Madison, Departments of Radiology and Medical Physics, Madison, WI 53705, USA
| | - Eduardo Aluicio-Sarduy
- University of Wisconsin-Madison, Departments of Radiology and Medical Physics, Madison, WI 53705, USA
| | - Todd E Barnhart
- University of Wisconsin-Madison, Departments of Radiology and Medical Physics, Madison, WI 53705, USA
| | - Jonathan W Engle
- University of Wisconsin-Madison, Departments of Radiology and Medical Physics, Madison, WI 53705, USA
- University of Wisconsin-Madison, Carbone Cancer Center, Madison, WI 53705, USA
| | - Weibo Cai
- University of Wisconsin-Madison, Department of Pharmaceutical Sciences, 600 Highland Ave., K6/562, Madison, WI 53792, USA
- University of Wisconsin-Madison, Departments of Radiology and Medical Physics, Madison, WI 53705, USA
- University of Wisconsin-Madison, Carbone Cancer Center, Madison, WI 53705, USA
| |
Collapse
|
15
|
Zhu Z, Liu Q, Zhu K, Wang K, Lin L, Chen Y, Shao F, Qian R, Song Y, Gao Y, Yang B, Jiang D, Lan X, An R. Aggregation-Induced Emission Photosensitizer/Bacteria Biohybrids Enhance Cerenkov Radiation-Induced Photodynamic Therapy by Activating Anti-Tumor Immunity for Synergistic Tumor Treatment. Acta Biomater 2023:S1742-7061(23)00334-3. [PMID: 37328041 DOI: 10.1016/j.actbio.2023.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/22/2023] [Accepted: 06/08/2023] [Indexed: 06/18/2023]
Abstract
Cerenkov radiation-induced photodynamic therapy (CR-PDT) gets rid of the limited tissue penetration depth of the external light source and provides a feasible scheme for the PDT excited by the internal light. However, due to the low luminescence intensity of Cerenkov radiation, CR-PDT alone cannot effectively inhibit tumor growth, curbing the potential clinical translation of CR-PDT. Herein, we reported an AIE-PS/bacteria biohybrid (EcN@TTVP) composed of Escherichia coli Nissle 1917 (EcN) loaded with aggregation-induced emission photosensitizer (AIE-PS) termed TTVP, which enhanced CR-PDT by activating anti-tumor immunity for synergistic tumor treatment. The preferential tumor-colonized EcN@TTVP and radiopharmaceutical 18F-fluorodeoxyglucose (18F-FDG) were administered sequentially to enable them to co-enrich in the tumor site, thereby triggering CR-PDT and promoting immunogenic tumor cell death. Most importantly, EcN acting as immunoadjuvants enhanced the maturation of dendritic cells (DCs) and priming of cytotoxic T cells (CTLs). Therefore, under the synergistic treatment of CR-PDT and immunotherapy, AIE-PS/bacteria biohybrids resulted in either efficient tumor remission or a survival prolongation in tumor-bearing mice, which presented significant advantages over single CR-PDT. Remarkably, no obvious toxic effects were observed during the treatment. In this study, we proposed a synergistic therapeutic strategy based on EcN@TTVP for combined CR-PDT and immunotherapy against tumors. Moreover, this strategy may have great potential in clinical translation and provide references for deep-seated tumor treatment. STATEMENT OF SIGNIFICANCE: PDT is restricted due to the shallow penetration depth of light into tumor tissues. Using CR as the excitation light source for PDT can overcome the aforementioned issue and greatly expand the application of PDT. However, the low efficacy of single CR-PDT limits further its applications. Therefore, the design and development of feasible strategies to improve the efficacy of CR-PDT are of immediate importance. Introducing probiotics to our study can be used not only as tumor-targeting carriers of photosensitizers but also as immunoadjuvants. Under co-stimulation by immunogenic tumor cell death triggered by CR-PDT and probiotics acting as immunoadjuvants, anti-tumor immune responses were effectively activated, thus remarkably enhancing the efficacy of CR-PDT.
Collapse
Affiliation(s)
- Ziyang Zhu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China; Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China; Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Ke Zhu
- Department of Nuclear Medicine, Zigong First People's Hospital, Zigong Academy of Medical Sciences, Zigong, 643000, China
| | - Kun Wang
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Lan Lin
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yaqi Chen
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Fuqiang Shao
- Department of Nuclear Medicine, Zigong First People's Hospital, Zigong Academy of Medical Sciences, Zigong, 643000, China
| | - Ruijie Qian
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China; Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Yangmeihui Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China; Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Yu Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China; Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Biao Yang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China; Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China; Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China; Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| | - Rui An
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China; Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| |
Collapse
|
16
|
Chen J, Zhou Y, Song M, Chen Y, Wang D, Huang Y, Hu P, He C, Dai T, Zhang L, Huang M, Chen Z, Xu P. A Serum-Stable supramolecular drug carrier for chemotherapeutics fabricated by a Peptide-Photosensitizer conjugate. J Colloid Interface Sci 2023; 646:959-969. [PMID: 37235941 DOI: 10.1016/j.jcis.2023.05.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/28/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023]
Abstract
Supramolecular assemblies fabricated by peptide-photosensitizer conjugates have attracted increasing attentions in recent years as drug carriers for chemotherapeutics (CTs). However, these assemblies have been known to suffer from disintegration by serum components leading to off-target drug release, and thereby impairing antitumor effects and causing systemic toxicities. To address this problem, this study reports a nano-architectural self-assembly peptide-photosensitizer carrier (NSPC) fabricated by conjugating a phthalocyanine derivative (MCPZnPc) and ε-poly-l-lysine (EPL). By engineering the core and peripheral interactions, MCPZnPC-EPL (M-E) NSPC firmly encapsulated multiple CTs, creating CT@M-E NSPCs that were highly stable against disintegration in serum. More importantly, CT@M-E NSPCs exhibited controlled release of CTs in tumor tissues. The antitumor effects of CTs were further promoted by the synergism with the reactivated photodynamic effect. Furthermore, M-E NSPC-encapsulation optimized CTs' biodistribution reducing adverse effects in vivo. This study provides a serum-stable supramolecular drug delivery system with photodynamic effect, which is applicable for a broad-range of CTs to promote antitumor effects and ameliorate adverse effects.
Collapse
Affiliation(s)
- Jincan Chen
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhou
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Meiru Song
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Yijian Chen
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, China
| | - Dong Wang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yunmei Huang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Ping Hu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, China
| | - Chen He
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Dai
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Zhuo Chen
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350116, China.
| |
Collapse
|
17
|
Tan Y, Yu D, Feng J, You H, Bai Y, He J, Cao H, Che Q, Guo J, Su Z. Toxicity evaluation of silica nanoparticles for delivery applications. Drug Deliv Transl Res 2023:10.1007/s13346-023-01312-z. [PMID: 37024610 DOI: 10.1007/s13346-023-01312-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2023] [Indexed: 04/08/2023]
Abstract
Silica nanoparticles (SiNPs) are being explored as nanocarriers for therapeutics delivery, which can address a number of intrinsic drawbacks of therapeutics. To translate laboratory innovation into clinical application, their potential toxicity has been of great concern. This review attempts to comprehensively summarize the existing literature on the toxicity assessment of SiNPs. The current data suggest that the composition of SiNPs, their physicochemical properties, their administration route, their frequency and duration of administration, and the sex of animal models are related to their tissue and blood toxicity, immunotoxicity, and genotoxicity. However, the correlation between in vitro and in vivo toxicity has not been well established, mainly because both the in vitro and the in vivo-dosed quantities are unrealistic. This article also discusses important factors to consider in the toxicology of SiNPs and current approaches to reducing their toxicity. The aim is to give readers a better understanding of the toxicology of silica nanoparticles and to help identify key gaps in knowledge and techniques.
Collapse
Affiliation(s)
- Yue Tan
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Dawei Yu
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jiayao Feng
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Huimin You
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510310, China
| | - Jincan He
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510310, China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, 528458, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd, Science City, Guangzhou, 510663, China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
18
|
Cerenkov radiation induced Chemo-Photodynamic Therapy using ROS-responsive agent. J Photochem Photobiol A Chem 2023. [DOI: 10.1016/j.jphotochem.2023.114641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
19
|
Choi PS, Lee JY, Chae JH, Wadas T, Cheng Z, Hur MG, Park JH. Theranostics through Utilizing Cherenkov Radiation of Radioisotope Zr-89 with a Nanocomposite Combination of TiO 2 and MnO 2. ACS APPLIED MATERIALS & INTERFACES 2023; 15:3689-3698. [PMID: 36573583 DOI: 10.1021/acsami.2c09195] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Cherenkov radiation (CR) derived from the decay of diagnostic and therapeutic radionuclides is currently being studied by the scientific community to determine if these emissions can be harnessed for cancer detection and therapy. While Cherenkov luminescence imaging (CLI) has been studied in the preclinical and clinical settings, Cherenkov radiation-induced cancer therapy (CRICT) is a relatively new area of research that harnesses the emitted photons to kill cancer cells through free radical generation and DNA damage. Nanoparticles seem well suited for developing a theranostic platform that would allow researchers to visualize therapy delivery and also generate the reactive oxygen species necessary to kill cancer cells. Herein, we report the preparation of an 89Zr-TiO2-MnO2 nanocomposite that incorporates transferrin onto the nanoparticle surface to enhance cancer cell growth inhibition. The incorporation of the positron emission tomography (PET) radioisotope 89Zr (half-life: 3.3 days) allowed for the detection of the nanoparticle using PET and for the creation of Cherenkov emissions that interacted with the nanoparticle surface to generate free radicals for therapy delivery. After preparation, these systems were observed to be stable in various media and provided excellent tumor growth control after being intratumorally injected into mice bearing CT-26 tumors. These results demonstrate that a therapeutically efficient CRICT platform can be generated using commercially available and affordable materials.
Collapse
Affiliation(s)
- Pyeong Seok Choi
- Accelerator Radioisotope Development Team, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do 56212, Republic of Korea
| | - Jun Young Lee
- Accelerator Radioisotope Development Team, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do 56212, Republic of Korea
| | - Jung Ho Chae
- Accelerator Radioisotope Development Team, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do 56212, Republic of Korea
| | - Thaddeus Wadas
- Department of Radiology, Carver College of Medicine, University of Iowa, 169 Newton Road, Iowa City, Iowa 52242, United States
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Min Goo Hur
- Radiation Utilization and Facilities Management Division, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do 56212, Republic of Korea
| | - Jeong Hoon Park
- Accelerator Radioisotope Development Team, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do 56212, Republic of Korea
| |
Collapse
|
20
|
Li X, Hsu JC, Son MH, Ha LN, Cai W. Cancer photodynamic therapy with chlorin e6-loaded, goat milk-derived extracellular vesicles: [ 18F]FDG lights up the way. Eur J Nucl Med Mol Imaging 2023; 50:247-250. [PMID: 36357594 PMCID: PMC9822859 DOI: 10.1007/s00259-022-06031-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Xiaoyan Li
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA
| | - Mai Hong Son
- Department of Nuclear Medicine, Hospital 108, Hanoi, Vietnam
| | - Le Ngoc Ha
- Department of Nuclear Medicine, Hospital 108, Hanoi, Vietnam
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of WI - Madison, Madison, WI, USA.
| |
Collapse
|
21
|
Guo R, Jiang D, Gai Y, Qian R, Zhu Z, Gao Y, Jing B, Yang B, Lan X, An R. Chlorin e6-loaded goat milk-derived extracellular vesicles for Cerenkov luminescence-induced photodynamic therapy. Eur J Nucl Med Mol Imaging 2023; 50:508-524. [PMID: 36222853 DOI: 10.1007/s00259-022-05978-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/16/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE Photodynamic therapy (PDT) is a promising cancer treatment strategy with rapid progress in preclinical and clinical settings. However, the limitations in penetration of external light and precise delivery of photosensitizers hamper its clinical translation. As such, the internal light source such as Cerenkov luminescence (CL) from decaying radioisotopes offers new opportunities. Herein, we show that goat milk-derived extracellular vesicles (GEV) can act as a carrier to deliver photosensitizer Chlorin e6 (Ce6) and tumor-avid 18F-FDG can activate CL-induced PDT for precision cancer theranostics. METHODS GEV was isolated via differential ultracentrifugation of commercial goat milk and photosensitizer Ce6 was loaded by co-incubation to obtain Ce6@GEV. Tumor uptake of Ce6@GEV was examined using confocal microscopy and flow cytometry. To demonstrate the ability of 18F-FDG to activate photodynamic effects against cancer cells, apoptosis rates were measured using flow cytometry, and the production of 1O2 was measured by reactive oxygen species (ROS) monitoring kit. Moreover, we used the IVIS device to detect Cherenkov radiation and Cerenkov radiation energy transfer (CRET). For animal experiments, a small-animal IVIS imaging system was used to visualize the accumulation of the GEV drug delivery system in tumors. PET/CT and CL images of the tumor site were performed at 0.5, 1, and 2 h. For in vivo antitumor therapy, changes of tumor volume, survival time, and body weight in six groups of tumor-bearing mice were monitored. Furthermore, the blood sample and organs of interest (heart, liver, spleen, lungs, kidneys, and tumor) were collected for hematological analysis, immunohistochemistry, and H&E staining. RESULTS Confocal microscopy of 4T1 cells incubated with Ce6@GEV for 4 h revealed strong red fluorescence signals in the cytoplasm, which demonstrated that Ce6 loaded in GEV could be efficiently delivered into tumor cells. When Ce6@GEV and 18F-FDG co-existed incubated with 4T1 cells, the cell viability plummeted from more than 88.02 ± 1.30% to 23.79 ± 1.59%, indicating excellent CL-induced PDT effects. In vivo fluorescence images showed a peak tumor/liver ratio of 1.36 ± 0.09 at 24 h after Ce6@GEV injection. For in vivo antitumor therapy, Ce6@GEV + 18F-FDG group had the best tumor inhibition rate (58.02%) compared with the other groups, with the longest survival rate (35 days, 40%). During the whole treatment process, neither blood biochemical analysis nor histological observation revealed vital organ damage, suggesting the biosafety of this treatment strategy. CONCLUSIONS The simultaneous accumulation of 18F-FDG and Ce6 in tumor tissues is expected to overcome the deficiency of traditional PDT. This strategy has the potential to extend PDT to a variety of tumors, including metastases, using targeted radiotracers to provide internal excitation of light-responsive therapeutics. We expect that our method will play a critical role in precision treatment of deep solid tumors.
Collapse
Affiliation(s)
- Rong Guo
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Ruijie Qian
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Ziyang Zhu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Yu Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Boping Jing
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Biao Yang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| | - Rui An
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| |
Collapse
|
22
|
Enhanced Photodynamic Therapy: A Review of Combined Energy Sources. Cells 2022; 11:cells11243995. [PMID: 36552759 PMCID: PMC9776440 DOI: 10.3390/cells11243995] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Photodynamic therapy (PDT) has been used in recent years as a non-invasive treatment for cancer, due to the side effects of traditional treatments such as surgery, radiotherapy, and chemotherapy. This therapeutic technique requires a photosensitizer, light energy, and oxygen to produce reactive oxygen species (ROS) which mediate cellular toxicity. PDT is a useful non-invasive therapy for cancer treatment, but it has some limitations that need to be overcome, such as low-light-penetration depths, non-targeting photosensitizers, and tumor hypoxia. This review focuses on the latest innovative strategies based on the synergistic use of other energy sources, such as non-visible radiation of the electromagnetic spectrum (microwaves, infrared, and X-rays), ultrasound, and electric/magnetic fields, to overcome PDT limitations and enhance the therapeutic effect of PDT. The main principles, mechanisms, and crucial elements of PDT are also addressed.
Collapse
|
23
|
Advanced techniques for performing photodynamic therapy in deep-seated tissues. Biomaterials 2022; 291:121875. [PMID: 36335717 DOI: 10.1016/j.biomaterials.2022.121875] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/07/2022] [Accepted: 10/23/2022] [Indexed: 11/23/2022]
Abstract
Photodynamic therapy (PDT) is a promising localized cancer treatment modality. It has been used successfully to treat a range of dermatological conditions with comparable efficacy to conventional treatments. However, some drawbacks limit the clinical utility of PDT in treating deep-seated tumors. Notably, the penetration limitation of UV and visible light, commonly applied to activate photosensitizers, makes PDT incompetent in treating deep-seated tumors. Development in light delivery technologies, especially fiber optics, led to improved clinical strategies for accessing deep tissues for irradiation. However, PDT efficacy issues remained partly due to light penetration limitations. In this review, we first summarized the current PDT applications for deep-seated tumor treatment. Then, the most recent progress in advanced techniques to overcome the light penetration limitation in PDT, including using functional nanomaterials that can either self-illuminate or be activated by near-infrared (NIR) light and X-rays as transducers, and implantable light delivery devices were discussed. Finally, current challenges and future opportunities of these technologies were discussed, which we hope may inspire the development of more effective techniques to enhance PDT efficacy against deep-seated tumors.
Collapse
|
24
|
Liu N, Su X, Sun X. Cerenkov radiation-activated probes for deep cancer theranostics: a review. Theranostics 2022; 12:7404-7419. [PMID: 36438500 PMCID: PMC9691350 DOI: 10.7150/thno.75279] [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: 05/19/2022] [Accepted: 09/07/2022] [Indexed: 11/05/2022] Open
Abstract
Cerenkov radiation (CR) from radionuclides and megavoltage X-ray radiation can act as an in situ light source for deep cancer theranostics, overcoming the limitations of external light sources. Despite the blue-weighted emission and low quantum yield of CR, activatable probes-mediated CR can enhance the in-vivo diagnostic signals by Cerenkov resonance energy transfer and also can produce therapeutic effects by reactive species generation/drug release, greatly promoting the biomedical applications of CR. In this review, we describe the principles and sources of CR, construction of CR-activated probes and their application to tumor optical imaging and therapy. Finally, future prospects for the design and biomedical application of CR-activated probes are discussed.
Collapse
Affiliation(s)
- Nian Liu
- PET Center, Department of Nuclear Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xinhui Su
- PET Center, Department of Nuclear Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaolian Sun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
25
|
Bianfei S, Fang L, Zhongzheng X, Yuanyuan Z, Tian Y, Tao H, Jiachun M, Xiran W, Siting Y, Lei L. Application of Cherenkov radiation in tumor imaging and treatment. Future Oncol 2022; 18:3101-3118. [PMID: 36065976 DOI: 10.2217/fon-2022-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cherenkov radiation (CR) is the characteristic blue glow that is generated during radiotherapy or radioisotope decay. Its distribution and intensity naturally reflect the actual dose and field of radiotherapy and the location of radioisotope imaging agents in vivo. Therefore, CR can represent a potential in situ light source for radiotherapy monitoring and radioisotope-based tumor imaging. When used in combination with new imaging techniques, molecular probes or nanomedicine, CR imaging exhibits unique advantages (accuracy, low cost, convenience and fast) in tumor radiotherapy monitoring and imaging. Furthermore, photosensitive nanomaterials can be used for CR photodynamic therapy, providing new approaches for integrating tumor imaging and treatment. Here the authors review the latest developments in the use of CR in tumor research and discuss current challenges and new directions for future studies.
Collapse
Affiliation(s)
- Shao Bianfei
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Liu Fang
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China.,Department of Radiation Oncology, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiang Zhongzheng
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Zeng Yuanyuan
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Tian
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - He Tao
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Ma Jiachun
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Wang Xiran
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Siting
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Liu Lei
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Malone CD, Egbulefu C, Zheleznyak A, Polina J, Karmakar P, Black K, Shokeen M, Achilefu S. Activation of nano-photosensitizers by Y-90 microspheres to enhance oxidative stress and cell death in hepatocellular carcinoma. Sci Rep 2022; 12:12748. [PMID: 35882949 PMCID: PMC9325688 DOI: 10.1038/s41598-022-17185-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/21/2022] [Indexed: 12/24/2022] Open
Abstract
While radioembolization with yttrium-90 (Y-90) microspheres is a promising treatment for hepatocellular carcinoma (HCC), lower responses in advanced and high-grade tumors present an urgent need to augment its tumoricidal efficacy. The purpose of this study was to determine whether clinically used Y-90 microspheres activate light-responsive nano-photosensitizers to enhance hepatocellular carcinoma (HCC) cell oxidative stress and cytotoxicity over Y-90 alone in vitro. Singlet oxygen and hydroxyl radical production was enhanced when Y-90 microspheres were in the presence of several nano-photosensitizers compared to either alone in cell-free conditions. Both the SNU-387 and HepG2 human HCC cells demonstrated significantly lower viability when treated with low activity Y-90 microspheres (0.1-0.2 MBq/0.2 mL) and a nano-photosensitizer consisting of both titanium dioxide (TiO2) and titanocene (TC) labelled with transferrin (TiO2-Tf-TC) compared to Y-90 microspheres alone or untreated cells. Cellular oxidative stress and cell death demonstrated a linear dependence on Y-90 at higher activities (up to 0.75 MBq/0.2 mL), but was significantly more accentuated in the presence of increasing TiO2-Tf-TC concentrations in the poorly differentiated SNU-387 HCC cell line (p < 0.0001 and p = 0.0002 respectively) but not the well-differentiated HepG2 cell line. Addition of TiO2-Tf-TC to normal human hepatocyte THLE-2 cells did not increase cellular oxidative stress or cell death in the presence of Y-90. The enhanced tumoricidal activity of nano-photosensitizers with Y-90 microspheres is a potentially promising adjunctive treatment strategy for certain patient subsets. Applications in clinically relevant in vivo HCC models are underway.
Collapse
Affiliation(s)
- Christopher D Malone
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Floor 2, St. Louis, MO, 63110, USA.
| | - Christopher Egbulefu
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Floor 2, St. Louis, MO, 63110, USA
| | - Alexander Zheleznyak
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Floor 2, St. Louis, MO, 63110, USA
| | - Jahnavi Polina
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Floor 2, St. Louis, MO, 63110, USA
| | - Partha Karmakar
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Floor 2, St. Louis, MO, 63110, USA
| | - Kvar Black
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Floor 2, St. Louis, MO, 63110, USA
| | - Monica Shokeen
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Floor 2, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Samuel Achilefu
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Floor 2, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
27
|
Nanodrug-loaded Bifidobacterium bifidum conjugated with anti-death receptor antibody for tumor-targeted photodynamic and sonodynamic synergistic therapy. Acta Biomater 2022; 146:341-356. [PMID: 35580829 DOI: 10.1016/j.actbio.2022.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022]
Abstract
Using bacteria for tumor-targeted therapy has attracted much attention in recent years. However, how to improve the targeted delivery and cancer therapy efficacy is an important but challenging scientific issue. Herein, a drug delivery system using a probiotic as a carrier was developed for tumor-targeted photodynamic and sonodynamic synergistic therapy. In this system, chlorin e6 (Ce6) nanoparticles (NPs) were prepared and incorporated into B. bifidum, followed by the conjugation of anti-death receptor 5 antibody (anti-DR5 Ab). Interestingly, B. bifidum under 671 nm laser or ultrasound (US) irradiation could generate reactive oxygen species (ROS), and Ce6-B. bifidum-anti-DR5 Ab obtained could target hypoxic regions in tumor with high efficiency after intravenous injection. The ROS level generated by Ce6-B. bifidum-anti-DR5 Ab under both laser and US irradiation was much higher than the combined ROS generated separately using a laser and US for the same probiotics. The cytotoxicity and laryngeal tumor growth-inhibiting efficiency of Ce6-B. bifidum-anti-DR5 Ab under both laser and US irradiation were significant higher than the values obtained using laser or US irradiation alone, which demonstrated the synergistic effect on tumor growth. B. bifidum could be eliminated from the body without exerting harmful effects on mouse health. This strategy is a platform that can be extended to treat other solid tumors. STATEMENT OF SIGNIFICANCE: Using bacteria as drug delivery carriers will show unique advantages. However, how to improve the targeted delivery efficiency and tumor inhibiting capacity is a challenging scientific issue. Herein, a delivery system using a probiotic as carrier was developed for tumor-targeted therapy. In this delivery system, chlorin e6 nanoparticles were prepared and then incorporated into living Bifidobacterium bifidum (B.bifidum), followed by the conjugation of anti-death receptor 5 antibody. This delivery system could efficiently target to mouse tumors, accumulate the hypoxic areas and inhibit the tumor growth through the photodynamic and sonodynamic synergistic effect. Our results will provide a platform for B.bifidum-mediated tumor targeted therapy.
Collapse
|
28
|
Abstract
Malignant tumors rank as a leading cause of death worldwide. Accurate diagnosis and advanced treatment options are crucial to win battle against tumors. In recent years, Cherenkov luminescence (CL) has shown its technical advantages and clinical transformation potential in many important fields, particularly in tumor diagnosis and treatment, such as tumor detection in vivo, surgical navigation, radiotherapy, photodynamic therapy, and the evaluation of therapeutic effect. In this review, we summarize the advances in CL for tumor diagnosis and treatment. We first describe the physical principles of CL and discuss the imaging techniques used in tumor diagnosis, including CL imaging, CL endoscope, and CL tomography. Then we present a broad overview of the current status of surgical resection, radiotherapy, photodynamic therapy, and tumor microenvironment monitoring using CL. Finally, we shed light on the challenges and possible solutions for tumor diagnosis and therapy using CL.
Collapse
|
29
|
Core-shell structured nanoparticles for photodynamic therapy-based cancer treatment and related imaging. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214427] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Qian R, Wang K, Guo Y, Li H, Zhu Z, Huang X, Gong C, Gao Y, Guo R, Yang B, Wang C, Jiang D, Lan X, An R, Gao Z. Minimizing adverse effects of Cerenkov radiation induced photodynamic therapy with transformable photosensitizer-loaded nanovesicles. J Nanobiotechnology 2022; 20:203. [PMID: 35477389 PMCID: PMC9044600 DOI: 10.1186/s12951-022-01401-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/28/2022] [Indexed: 01/12/2023] Open
Abstract
Background Photodynamic therapy (PDT) is a promising antitumor strategy with fewer adverse effects and higher selectivity than conventional therapies. Recently, a series of reports have suggested that PDT induced by Cerenkov radiation (CR) (CR-PDT) has deeper tissue penetration than traditional PDT; however, the strategy of coupling radionuclides with photosensitizers may cause severe side effects. Methods We designed tumor-targeting nanoparticles (131I-EM@ALA) by loading 5-aminolevulinic acid (ALA) into an 131I-labeled exosome mimetic (EM) to achieve combined antitumor therapy. In addition to playing a radiotherapeutic role, 131I served as an internal light source for the Cerenkov radiation (CR). Results The drug-loaded nanoparticles effectively targeted tumors as confirmed by confocal imaging, flow cytometry, and small animal fluorescence imaging. In vitro and in vivo experiments demonstrated that 131I-EM@ALA produced a promising antitumor effect through the synergy of radiotherapy and CR-PDT. The nanoparticles killed tumor cells by inducing DNA damage and activating the lysosome-mitochondrial pathways. No obvious abnormalities in the hematology analyses, blood biochemistry, or histological examinations were observed during the treatment. Conclusions We successfully engineered a nanocarrier coloaded with the radionuclide 131I and a photosensitizer precursor for combined radiotherapy and PDT for the treatment of breast cancer. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01401-0.
Collapse
Affiliation(s)
- Ruijie Qian
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Kun Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yawen Guo
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Ziyang Zhu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiaojuan Huang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Department of Nuclear Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Chengpeng Gong
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yu Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Rong Guo
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Biao Yang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chenyang Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Rui An
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China. .,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Zairong Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China. .,Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
31
|
Roy I, Krishnan S, Kabashin AV, Zavestovskaya IN, Prasad PN. Transforming Nuclear Medicine with Nanoradiopharmaceuticals. ACS NANO 2022; 16:5036-5061. [PMID: 35294165 DOI: 10.1021/acsnano.1c10550] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nuclear medicine is expected to make major advances in cancer diagnosis and therapy; tumor-targeted radiopharmaceuticals preferentially eradicate tumors while causing minimal damage to healthy tissues. The current scope of nuclear medicine can be significantly expanded by integration with nanomedicine, which utilizes nanoparticles for cancer diagnosis and therapy by capitalizing on the increased surface area-to-volume ratio, the passive/active targeting ability and high loading capacity, the greater interaction cross section with biological tissues, the rich surface properties of nanomaterials, the facile decoration of nanomaterials with a plethora of functionalities, and the potential for multiplexing several functionalities within one construct. This review provides a comprehensive discussion of nuclear nanomedicine using tumor-targeted nanoparticles for cancer radiation therapy with either pre-embedded radionuclides or nonradioactive materials which can be extrinsically triggered using various external nuclear particle sources to produce in situ radioactivity. In addition, it describes the prospect of combining nuclear nanomedicine with other modalities to enable synergistically enhanced combination therapies. The review also discusses advances in the fabrication of radionuclides as well as describes laser ablation technologies for producing nanoradiopharmaceuticals, which combine the ease of production with exceptional purity and rapid biodegradability, along with additional imaging or therapeutic functionalities. From a practical standpoint, these attributes of nanoradiopharmaceuticals may provide distinct advantages in diagnostic/therapeutic sensitivity and specificity, imaging resolution, and scalability of turnkey platforms. Coupling image-guided targeted radiation therapy with the possibility of in situ activation of nanomaterials as well as combining with other therapeutic modalities using a multifunctional nanoplatform could herald an era of exciting technological and therapeutic advances to radically transform the landscape of nuclear medicine. The review concludes with a discussion of current challenges and presents the authors' views on future opportunities to stimulate further research in this rewarding field of high societal impact.
Collapse
Affiliation(s)
- Indrajit Roy
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Andrei V Kabashin
- Aix Marseille University, CNRS, LP3, Campus de Luminy - Case 917, 13288 Marseille, France
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
| | - Irina N Zavestovskaya
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
- Nuclear Physics and Astrophysics Department, LPI of RAS, 119991 Moscow, Russia
| | - Paras N Prasad
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), 115409 Moscow, Russia
- Department of Chemistry and Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| |
Collapse
|
32
|
Deep-Tissue Activation of Photonanomedicines: An Update and Clinical Perspectives. Cancers (Basel) 2022; 14:cancers14082004. [PMID: 35454910 PMCID: PMC9032169 DOI: 10.3390/cancers14082004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Photodynamic therapy (PDT) is a light-activated treatment modality, which is being clinically used and further developed for a number of premalignancies, solid tumors, and disseminated cancers. Nanomedicines that facilitate PDT (photonanomedicines, PNMs) have transformed its safety, efficacy, and capacity for multifunctionality. This review focuses on the state of the art in deep-tissue activation technologies for PNMs and explores how their preclinical use can evolve towards clinical translation by harnessing current clinically available instrumentation. Abstract With the continued development of nanomaterials over the past two decades, specialized photonanomedicines (light-activable nanomedicines, PNMs) have evolved to become excitable by alternative energy sources that typically penetrate tissue deeper than visible light. These sources include electromagnetic radiation lying outside the visible near-infrared spectrum, high energy particles, and acoustic waves, amongst others. Various direct activation mechanisms have leveraged unique facets of specialized nanomaterials, such as upconversion, scintillation, and radiosensitization, as well as several others, in order to activate PNMs. Other indirect activation mechanisms have leveraged the effect of the interaction of deeply penetrating energy sources with tissue in order to activate proximal PNMs. These indirect mechanisms include sonoluminescence and Cerenkov radiation. Such direct and indirect deep-tissue activation has been explored extensively in the preclinical setting to facilitate deep-tissue anticancer photodynamic therapy (PDT); however, clinical translation of these approaches is yet to be explored. This review provides a summary of the state of the art in deep-tissue excitation of PNMs and explores the translatability of such excitation mechanisms towards their clinical adoption. A special emphasis is placed on how current clinical instrumentation can be repurposed to achieve deep-tissue PDT with the mechanisms discussed in this review, thereby further expediting the translation of these highly promising strategies.
Collapse
|
33
|
Brevé TG, Filius M, Weerdenburg S, van der Griend SJ, Groeneveld TP, Denkova AG, Eelkema R. Light-Sensitive Phenacyl Crosslinked Dextran Hydrogels for Controlled Delivery. Chemistry 2022; 28:e202103523. [PMID: 34939694 PMCID: PMC9306828 DOI: 10.1002/chem.202103523] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Indexed: 11/16/2022]
Abstract
Stimuli-responsive soft materials enable controlled release of loaded drug molecules and biomolecules. Controlled release of potent chemotherapeutic or immunotherapeutic agents is crucial to reduce unwanted side effects. In an effort to develop controlled release strategies that can be triggered by using Cerenkov luminescence, we have developed polymer hydrogels that can release bovine serum albumin and immunoglobulin G by using light (254 nm-375 nm) as a trigger. We describe the synthesis and photochemical characterization of two light sensitive phenacyl bis-azide crosslinkers that are used to prepare transparent self-supporting hydrogel patches. One crosslinker was designed to optimize the overlap with the Cerenkov luminescence emission window, bearing an π-extended phenacyl core, resulting in a high quantum yield (14 %) of photocleavage when irradiated with 375 nm light. We used the extended phenacyl crosslinker for the preparation of protein-loaded dextran hydrogel patches, which showed efficient and selective dosed release of bovine serum albumin or immunoglobulin G after irradiation with 375 nm light. Cerenkov-triggered release is as yet inconclusive due to unexpected side-reactivity. Based on the high quantum yield, efficient release and large overlap with the Cerenkov window, we envision application of these photosensitive soft materials in radiation targeted drug release.
Collapse
Affiliation(s)
- Tobias G. Brevé
- Department of Chemical EngineeringDelft University of Technologyvan der Maasweg 92629 HZDelftThe Netherlands
| | - Mike Filius
- Department of BioNanoScienceDelft University of Technologyvan der Maasweg 92629 HZDelftThe Netherlands
| | - Sven Weerdenburg
- Department of Chemical EngineeringDelft University of Technologyvan der Maasweg 92629 HZDelftThe Netherlands
| | - Stefan J. van der Griend
- Department of Chemical EngineeringDelft University of Technologyvan der Maasweg 92629 HZDelftThe Netherlands
| | - Tim P. Groeneveld
- Department of Chemical EngineeringDelft University of Technologyvan der Maasweg 92629 HZDelftThe Netherlands
| | - Antonia G. Denkova
- Department of Radiation Science and TechnologyDelft University of TechnologyMekelweg 152629 JBDelftThe Netherlands
| | - Rienk Eelkema
- Department of Chemical EngineeringDelft University of Technologyvan der Maasweg 92629 HZDelftThe Netherlands
| |
Collapse
|
34
|
Tiwari AK, Mishra A, Pandey G, Gupta MK, Pandey PC. Nanotechnology: A Potential Weapon to Fight against COVID-19. PARTICLE & PARTICLE SYSTEMS CHARACTERIZATION : MEASUREMENT AND DESCRIPTION OF PARTICLE PROPERTIES AND BEHAVIOR IN POWDERS AND OTHER DISPERSE SYSTEMS 2022; 39:2100159. [PMID: 35440846 PMCID: PMC9011707 DOI: 10.1002/ppsc.202100159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/28/2021] [Indexed: 05/13/2023]
Abstract
The COVID-19 infections have posed an unprecedented global health emergency, with nearly three million deaths to date, and have caused substantial economic loss globally. Hence, an urgent exploration of effective and safe diagnostic/therapeutic approaches for minimizing the threat of this highly pathogenic coronavirus infection is needed. As an alternative to conventional diagnosis and antiviral agents, nanomaterials have a great potential to cope with the current or even future health emergency situation with a wide range of applications. Fundamentally, nanomaterials are physically and chemically tunable and can be employed for the next generation nanomaterial-based detection of viral antigens and host antibodies in body fluids as antiviral agents, nanovaccine, suppressant of cytokine storm, nanocarrier for efficient delivery of antiviral drugs at infection site or inside the host cells, and can also be a significant tool for better understanding of the gut microbiome and SARS-CoV-2 interaction. The applicability of nanomaterial-based therapeutic options to cope with the current and possible future pandemic is discussed here.
Collapse
Affiliation(s)
- Atul K. Tiwari
- Department of ChemistryIndian Institute of Technology (BHU)VaranasiUttar Pradesh221005India
| | - Anupa Mishra
- Department of MicrobiologyDr. R.M.L. Awadh UniversityAyodhyaUttar Pradesh224001India
- Department of MicrobiologySri Raghukul Mahila Vidya PeethCivil Line GondaUttar Pradesh271001India
| | - Govind Pandey
- Department of PaediatricsKing George Medical UniversityLucknowUttar Pradesh226003India
| | - Munesh K. Gupta
- Department of MicrobiologyInstitute of Medical SciencesBanaras Hindu UniversityVaranasiUttar Pradesh221005India
| | - Prem C. Pandey
- Department of ChemistryIndian Institute of Technology (BHU)VaranasiUttar Pradesh221005India
| |
Collapse
|
35
|
Zheng Y, Li Z, Yang Y, Shi H, Chen H, Gao Y. A nanosensitizer self-assembled from oleanolic acid and chlorin e6 for synergistic chemo/sono-photodynamic cancer therapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 93:153788. [PMID: 34634745 DOI: 10.1016/j.phymed.2021.153788] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/25/2021] [Accepted: 09/29/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Sono-photodynamic therapy (SPDT) which is the combination of photodynamic therapy (PDT) and sonodynamic therapy (SDT), could exert much better anti-cancer effects than monotherapy. The combination of chemotherapy and PDT or SDT has shown great potential for cancer treatment. However, the combination of SPDT and chemotherapy for cancer treatment is rarely explored. PURPOSE We utilized a natural hydrophobic anti-cancer drug oleanolic acid (OA) and a photosensitizer chlorin e6 (Ce6) through self-assembly technology to form a carrier-free nanosensitizer OC for combined chemotherapy and SPDT for cancer treatment. No studies involving using carrier-free nanomedicine for combined chemotherapy/SPDT have been reported yet. STUDY DESIGN After fully characterization of OC, the in vitro and in vivo anti-cancer activities of OC were investigated and the mechanisms of the synergistic therapeutic effects were studied. METHODS OC were synthesized through self-assembly technology and characterized by dynamic light scattering (DLS) and an atomic force microscope (AFM). Confocal microscope was used to investigate the intracellular uptake efficiency and the penetration ability of OC. The cell viability of PC9 and 4T1 cells treated with OC under laser and ultrasound (US) irradiation was determined by MTT assay. Furthermore, flow cytometry was performed to detect the reactive oxygen species (ROS) generation, loss of mitochondrial membrane potential (MMP), cell apoptosis and cell cycle arrest. Finally, the anti-tumor therapeutic efficacy of OC was investigated in orthotopic 4T1 breast tumor-bearing mouse model. RESULTS OC showed an average particle size of around 100 nm with excellent light stability. OC increased more than 23 times accumulation of Ce6 in cancer cells and had strong tumor penetration ability in three-dimensional (3D) multicellular tumor spheroids (MCTSs). Compared with other therapeutic options, OC showed obvious synergistic inhibitory effects under light and US irradiation in PC9 and 4T1 cells with a significant decrease in IC50 values. Mechanism studies showed that OC could generate high ROS, induce MMP loss, and cause apoptosis and cell cycle arrest. In vivo studies also approved the synergistic therapeutic effects of OC in 4T1 mouse models. CONCLUSION Self-assembled carrier-free nanosensitizer OC could be a promising therapeutic agent for synergistic chemo/sono-photodynamic therapy for cancer treatment.
Collapse
Affiliation(s)
- Yilin Zheng
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Ziying Li
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Ya Yang
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Huifang Shi
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Haijun Chen
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Yu Gao
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China.
| |
Collapse
|
36
|
Li J, Dai S, Qin R, Shi C, Ming J, Zeng X, Wen X, Zhuang R, Chen X, Guo Z, Zhang X. Ligand Engineering of Titanium-Oxo Nanoclusters for Cerenkov Radiation-Reinforced Photo/Chemodynamic Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:54727-54738. [PMID: 34766763 DOI: 10.1021/acsami.1c16213] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The therapeutic effect of general photodynamic therapy (PDT) is gravely limited by the poor penetration depth of exogenous light radiation. In recent years, Cerenkov radiation (CR) has been exploringly applied to overcome this critical defect. However, the currently reported type I photosensitizers for CR-induced PDT (CRIT) are only TiO2 nanoparticle-based agents with numerous fatally intrinsic drawbacks. Herein, we developed NH2-Ti32O16 nanocluster (NTOC)-derived ultrasmall nanophotosensitizers (NPSs, denoted as TDPs) via innovate ligand engineering. The introduced dopamine (DA) ligands not only facilitate the water solubility and photocatalytic properties of NPSs but also involve the tumor-targeting behavior through the binding affinity with DA receptors on cancer cells. Under CR irradiation, TDPs enable efficient hydroxyl radical (·OH) generation benefiting from the enhanced separation of hole (h+)-electron (e-) pairs, where the h+ will react with H2O to execute type I PDT and the transferred e- can realize the augmentation of Ti3+ to substantially promote the therapeutic index of chemodynamic therapy. This study provides an easy but feasible strategy for constructing versatile NPSs with an ultrasmall framework structure, propounding a refreshing paradigm for implementing efficient CR-induced combined therapy (CRICT) and spurring the development of CR and titanium-familial nanoplatforms in the fields of photocatalysis and nanocatalytic medicine.
Collapse
Affiliation(s)
- Jingchao Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Shuqi Dai
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, People's Republic of China
| | - Ruixue Qin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Changrong Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jiang Ming
- Department of Chemistry, Fudan University, Shanghai 200438, People's Republic of China
| | - Xinying Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xuejun Wen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Rongqiang Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore 117597, Singapore
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
37
|
Yang YL, Lin K, Yang L. Progress in Nanocarriers Codelivery System to Enhance the Anticancer Effect of Photodynamic Therapy. Pharmaceutics 2021; 13:1951. [PMID: 34834367 PMCID: PMC8617654 DOI: 10.3390/pharmaceutics13111951] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 02/05/2023] Open
Abstract
Photodynamic therapy (PDT) is a promising anticancer noninvasive method and has great potential for clinical applications. Unfortunately, PDT still has many limitations, such as metastatic tumor at unknown sites, inadequate light delivery and a lack of sufficient oxygen. Recent studies have demonstrated that photodynamic therapy in combination with other therapies can enhance anticancer effects. The development of new nanomaterials provides a platform for the codelivery of two or more therapeutic drugs, which is a promising cancer treatment method. The use of multifunctional nanocarriers for the codelivery of two or more drugs can improve physical and chemical properties, increase tumor site aggregation, and enhance the antitumor effect through synergistic actions, which is worthy of further study. This review focuses on the latest research progress on the synergistic enhancement of PDT by simultaneous multidrug administration using codelivery nanocarriers. We introduce the design of codelivery nanocarriers and discuss the mechanism of PDT combined with other antitumor methods. The combination of PDT and chemotherapy, gene therapy, immunotherapy, photothermal therapy, hyperthermia, radiotherapy, sonodynamic therapy and even multidrug therapy are discussed to provide a comprehensive understanding.
Collapse
Affiliation(s)
| | | | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.-L.Y.); (K.L.)
| |
Collapse
|
38
|
Ávila-Sánchez MA, Isaac-Olivé K, Aranda-Lara L, Morales-Ávila E, Plata-Becerril A, Jiménez-Mancilla NP, Ocampo-García B, Estrada JA, Santos-Cuevas CL, Torres-García E, Camacho-López MA. Targeted photodynamic therapy using reconstituted high-density lipoproteins as rhodamine transporters. Photodiagnosis Photodyn Ther 2021; 37:102630. [PMID: 34798347 DOI: 10.1016/j.pdpdt.2021.102630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 10/09/2021] [Accepted: 11/12/2021] [Indexed: 01/10/2023]
Abstract
Reconstituted high-density lipoprotein (rHDL) nanoparticles are excellent transporters of molecules and very useful for targeted therapy as they specifically recognize the scavenger receptor, class B1 (SR-B1) that is present on the surface of a wide range of tumor cells. However, they have rarely been employed to transport photosensitizers (PS) for photodynamic therapy (PDT). Rhodamine (R) compounds have been dismissed as useful PSs for PDT due to their low 1O2 production, excitation wavelengths with little tissue penetration, and poor selectivity for tumor cells. It was recently demonstrated that when irradiating at 532 nm or with Cerenkov radiation (CR) from a β-emitting radionuclide, R123, R6G, and RB undergo electron transfer reactions (type I reaction) with folic acid. R6G also produces type I reactions with O2. In this work, the photodynamic effects of the rHDL-R system were evaluated in vitro. rHDL nanoparticles loaded with R123, R6G, and RB were synthesized, and the PS was internalized into T47D tumor cells. When cells were irradiated with a 532-nm laser in the presence of an rHDL-R systems, a cytotoxic photodynamic effect was obtained in the order R6G > R123 > RB. In the presence of CR from a 177Lu source, cytotoxicity showed the order R6G > RB > R123. The higher cytotoxicity induced by R6G in both cases corresponds to higher cellular internalization and larger production of type I and II reactions. Thus, in this work, it is proposed that rHDL-R/177Lu system can be applied in theragnostics as a multimodal radiotherapy-PDT-imaging system (imaging by SPECT or Cerenkov) and in hypoxic solid tumors in which external radiation is not effective and 177Lu-CR acts as light source.
Collapse
Affiliation(s)
- Marcela A Ávila-Sánchez
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México 50180, Mexico
| | - Keila Isaac-Olivé
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México 50180, Mexico.
| | - Liliana Aranda-Lara
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México 50180, Mexico.
| | - Enrique Morales-Ávila
- Laboratorio de Toxicología y Farmacia, Facultad de Química, Universidad Autónoma del Estado de México, Toluca, Estado de México 50120, Mexico
| | - Adriana Plata-Becerril
- Laboratorio de Toxicología y Farmacia, Facultad de Química, Universidad Autónoma del Estado de México, Toluca, Estado de México 50120, Mexico
| | - Nallely P Jiménez-Mancilla
- Laboratorio Nacional de Investigación y Desarrollo de Radiofármacos-CONACyT, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México 52750, Mexico.
| | - Blanca Ocampo-García
- Laboratorio Nacional de Investigación y Desarrollo de Radiofármacos-CONACyT, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México 52750, Mexico
| | - José A Estrada
- Laboratorio de Neuroquímica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México 50180, Mexico
| | - Clara L Santos-Cuevas
- Laboratorio Nacional de Investigación y Desarrollo de Radiofármacos-CONACyT, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México 52750, Mexico
| | - Eugenio Torres-García
- Laboratorio de Dosimetría y Simulación Monte Carlo, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México 50180, Mexico
| | - Miguel A Camacho-López
- Laboratorio de Fotomedicina, Biofotónica y Espectroscopía Láser de Pulsos Ultracortos, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México 50180, Mexico
| |
Collapse
|
39
|
Lin L, Song X, Dong X, Li B. Nano-photosensitizers for enhanced photodynamic therapy. Photodiagnosis Photodyn Ther 2021; 36:102597. [PMID: 34699982 DOI: 10.1016/j.pdpdt.2021.102597] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 12/22/2022]
Abstract
Photodynamic therapy (PDT) utilizes photosensitizers (PSs) together with irradiation light of specific wavelength interacting with oxygen to generate cytotoxic reactive oxygen species (ROS), which could trigger apoptosis and/or necrosis-induced cell death in target tissues. During the past two decades, multifunctional nano-PSs employing nanotechnology and nanomedicine developed, which present not only photosensitizing properties but additionally accurate drug release abilities, efficient response to optical stimuli and hypoxia resistance. Further, nano-PSs have been developed to enhance PDT efficacy by improving the ROS yield. In addition, nano-PSs with additive or synergistic therapies are significant for both currently preclinical study and future clinical practice, given their capability of considerable higher therapeutic efficacy under safer systemic drug dosage. In this review, nano-PSs that allow precise drug delivery for efficient absorption by target cells are introduced. Nano-PSs boosting sensitivity and conversion efficiency to PDT-activating stimuli are highlighted. Nano-PSs developed to address the challenging hypoxia conditions during PDT of deep-sited tumors are summarized. Specifically, PSs capable of synergistic therapy and the emerging novel types with higher ROS yield that further enhance PDT efficacy are presented. Finally, future demands for ideal nano-PSs, emphasizing clinical translation and application are discussed.
Collapse
Affiliation(s)
- Li Lin
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, 350117, China
| | - Xuejiao Song
- Key Laboratory of Flexible Electronics and Institute of Advanced Materials, Nanjing Technology University, Nanjing 211800, China
| | - Xiaocheng Dong
- Key Laboratory of Flexible Electronics and Institute of Advanced Materials, Nanjing Technology University, Nanjing 211800, China
| | - Buhong Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, 350117, China.
| |
Collapse
|
40
|
Guo J, Feng K, Wu W, Ruan Y, Liu H, Han X, Shao G, Sun X. Smart
131
I‐Labeled Self‐Illuminating Photosensitizers for Deep Tumor Therapy. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202107231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jingru Guo
- State Key Laboratory of Natural Medicines Key Laboratory of Drug Quality Control and Pharmacovigilance Department of Pharmaceutical Analysis China Pharmaceutical University Nanjing 210009 China
| | - Kai Feng
- State Key Laboratory of Natural Medicines Key Laboratory of Drug Quality Control and Pharmacovigilance Department of Pharmaceutical Analysis China Pharmaceutical University Nanjing 210009 China
| | - Wenyu Wu
- Department of Nuclear Medicine Nanjing First Hospital Nanjing Medical University Nanjing 210006 China
| | - Yiling Ruan
- State Key Laboratory of Natural Medicines Key Laboratory of Drug Quality Control and Pharmacovigilance Department of Pharmaceutical Analysis China Pharmaceutical University Nanjing 210009 China
| | - Huihui Liu
- State Key Laboratory of Natural Medicines Key Laboratory of Drug Quality Control and Pharmacovigilance Department of Pharmaceutical Analysis China Pharmaceutical University Nanjing 210009 China
| | - Xiuping Han
- Department of Nuclear Medicine Nanjing First Hospital Nanjing Medical University Nanjing 210006 China
| | - Guoqiang Shao
- Department of Nuclear Medicine Nanjing First Hospital Nanjing Medical University Nanjing 210006 China
| | - Xiaolian Sun
- State Key Laboratory of Natural Medicines Key Laboratory of Drug Quality Control and Pharmacovigilance Department of Pharmaceutical Analysis China Pharmaceutical University Nanjing 210009 China
| |
Collapse
|
41
|
Guo J, Feng K, Wu W, Ruan Y, Liu H, Han X, Shao G, Sun X. Smart 131 I-Labeled Self-Illuminating Photosensitizers for Deep Tumor Therapy. Angew Chem Int Ed Engl 2021; 60:21884-21889. [PMID: 34374188 DOI: 10.1002/anie.202107231] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Indexed: 12/14/2022]
Abstract
Stimulating photosensitizers (PS) by Cerenkov radiation (CR) can overcome the light penetration limitation in traditional photodynamic therapy. However, separate injection of radiopharmaceuticals and PS cannot guarantee their efficient interaction in tumor areas, while co-delivery of radionuclides and PS face the problem of nonnegligible phototoxicity in normal tissues. Here, we describe a 131 I-labeled smart photosensitizer, composed of pyropheophorbide-a (photosensitizer), a diisopropylamino group (pH-sensitive group), an 131 I-labeled tyrosine group (CR donor), and polyethylene glycol, which can self-assemble into nanoparticles (131 I-sPS NPs). The 131 I-sPS NPs showed low phototoxicity in normal tissues due to aggregation-caused quenching effect, but could self-produce reactive oxygen species in tumor sites upon disassembly. Upon intravenous injection, 131 I-sPS NPs showed great tumor inhibition capability in subcutaneous 4T1-tumor-bearing Balb/c mice and orthotopic VX2 liver tumor bearing rabbits. We believed 131 I-sPS NPs could expand the application of CR and provide an effective strategy for deep tumor theranostics.
Collapse
Affiliation(s)
- Jingru Guo
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Kai Feng
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Wenyu Wu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yiling Ruan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Huihui Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiuping Han
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Guoqiang Shao
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaolian Sun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
42
|
Song F, Li S, Sun C, Ji Y, Zhang Y. ROS-Responsive Selenium-Containing Carriers for Coencapsulation of Photosensitizer and Hypoxia-Activated Prodrug and Their Cellular Behaviors. Macromol Biosci 2021; 21:e2100229. [PMID: 34390189 DOI: 10.1002/mabi.202100229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/06/2021] [Indexed: 11/08/2022]
Abstract
The integration of hypoxia-activated chemotherapy with photodynamic therapy (PDT) has newly become a potent strategy for tumor treatment. Herein, a reactive oxygen species (ROS)-responsive drug carriers (PS@AQ4N/mPEG-b-PSe NPs) are fabricated based on the amphiphilic selenium-containing methoxy poly(ethylene glycol)-polycarbonate (mPEG-b-PSe), the hydrophobic photosensitizer (PS), and hypoxia-activated prodrug Banoxantrone (AQ4N). The obtained nanoparticles are spherical with an average diameter of 100 nm as characterized by transmission electron microscope (TEM) and dynamic laser scattering (DLS) respectively. The encapsulation efficiency of the PS and AQ4N reaches 92.83% and 51.04% at different conditions, respectively, by UV-vis spectrophotometer. It is found that the drug release is accelerated due to the good ROS responsiveness of mPEG-b-PSe and the cumulative release of AQ4N is up to 89% within 30 h. The cell test demonstrates that the nanoparticles dissociate when triggered by the ROS stimuli in the cancer cells, thus the PS is exposed to more oxygen and the ROS generation efficiency is enhanced accordingly. The consumption of oxygen during PDT leads to the increased tumor hypoxia, and subsequently activates AQ4N into cytotoxic counterpart to inhibit tumor growth. Therefore, the synergistic therapeutic efficacy demonstrates this drug delivery has great potential for antitumor therapy.
Collapse
Affiliation(s)
- Fangqin Song
- Shanghai Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials and Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Siqi Li
- Shanghai Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials and Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Chuanhao Sun
- Shanghai Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials and Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Ying Ji
- Institute of Textiles and Clothing, Hong Kong Polytechnic University, Hunghom, Kowloon, Hong Kong SAR, 999077, China
| | - Yan Zhang
- Shanghai Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials and Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China.,Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Shanghai, 201203, China
| |
Collapse
|
43
|
Theranostic Applications of Nanoparticle-Mediated Photoactivated Therapies. JOURNAL OF NANOTHERANOSTICS 2021. [DOI: 10.3390/jnt2030009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nanoparticle-mediated light-activated therapies, such as photodynamic therapy and photothermal therapy, are earnestly being viewed as efficient interventional strategies against several cancer types. Theranostics is a key hallmark of cancer nanomedicine since it allows diagnosis and therapy of both primary and metastatic cancer using a single nanoprobe. Advanced in vivo diagnostic imaging using theranostic nanoparticles not only provides precise information about the location of tumor/s but also outlines the narrow time window corresponding to the maximum tumor-specific drug accumulation. Such information plays a critical role in guiding light-activated therapies with high spatio-temporal accuracy. Furthermore, theranostics facilitates monitoring the progression of therapy in real time. Herein, we provide a general review of the application of theranostic nanoparticles for in vivo image-guided light-activated therapy in cancer. The imaging modalities considered here include fluorescence imaging, photoacoustic imaging, thermal imaging, magnetic resonance imaging, X-ray computed tomography, positron emission tomography, and single-photon emission computed tomography. The review concludes with a brief discussion about the broad scope of theranostic light-activated nanomedicine.
Collapse
|
44
|
Bayoumi NA, El-Kolaly MT. Utilization of nanotechnology in targeted radionuclide cancer therapy: monotherapy, combined therapy and radiosensitization. RADIOCHIM ACTA 2021. [DOI: 10.1515/ract-2020-0098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abstract
The rapid progress of nanomedicine field has a great influence on the different tumor therapeutic trends. It achieves a potential targeting of the therapeutic agent to the tumor site with neglectable exposure of the normal tissue. In nuclear medicine, nanocarriers have been employed for targeted delivery of therapeutic radioisotopes to the malignant tissues. This systemic radiotherapy is employed to overcome the external radiation therapy drawbacks. This review overviews studies concerned with investigation of different nanoparticles as promising carriers for targeted radiotherapy. It discusses the employment of different nanovehicles for achievement of the synergistic effect of targeted radiotherapy with other tumor therapeutic modalities such as hyperthermia and photodynamic therapy. Radiosensitization utilizing different nanosensitizer loaded nanoparticles has also been discussed briefly as one of the nanomedicine approach in radiotherapy.
Collapse
Affiliation(s)
- Noha Anwer Bayoumi
- Department of Radiolabeled Compounds , Hot Laboratories Center, Egyptian Atomic Energy Authority , Cairo , Egypt
| | - Mohamed Taha El-Kolaly
- Department of Radiolabeled Compounds , Hot Laboratories Center, Egyptian Atomic Energy Authority , Cairo , Egypt
| |
Collapse
|
45
|
Zhong X, Wang X, Li J, Hu J, Cheng L, Yang X. ROS-based dynamic therapy synergy with modulating tumor cell-microenvironment mediated by inorganic nanomedicine. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213828] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
46
|
Chen YA, Li JJ, Lin SL, Lu CH, Chiu SJ, Jeng FS, Chang CW, Yang BH, Chang MC, Ke CC, Liu RS. Effect of Cerenkov Radiation-Induced Photodynamic Therapy with 18F-FDG in an Intraperitoneal Xenograft Mouse Model of Ovarian Cancer. Int J Mol Sci 2021; 22:4934. [PMID: 34066508 PMCID: PMC8125334 DOI: 10.3390/ijms22094934] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer (OC) metastases frequently occur through peritoneal dissemination, and they contribute to difficulties in treatment. While photodynamic therapy (PDT) has the potential to treat OC, its use is often limited by tissue penetration depth and tumor selectivity. Herein, we combined Cerenkov radiation (CR) emitted by 18F-FDG accumulated in tumors as an internal light source and several photosensitizer (PS) candidates with matched absorption bands, including Verteporfin (VP), Chlorin e6 (Ce6) and 5'-Aminolevulinic acid (5'-ALA), to evaluate the anti-tumor efficacy. The in vitro effect of CR-induced PDT (CR-PDT) was evaluated using a cell viability assay, and the efficiency of PS was assessed by measuring the singlet oxygen production. An intraperitoneal ES2 OC mouse model was used for in vivo evaluation of CR-PDT. Positron emission tomography (PET) imaging and bioluminescence-based imaging were performed to monitor the biologic uptake of 18F-FDG and the therapeutic effect. The in vitro studies demonstrated Ce6 and VP to be more effective PSs for CR-PDT. Moreover, VP was more efficient in the generation of singlet oxygen and continued for a long time when exposed to fluoro-18 (18F). Combining CR emitted by 18F-FDG and VP treatment not only significantly suppressed tumor growth, but also prolonged median survival times compared to either monotherapy.
Collapse
Affiliation(s)
- Yi-An Chen
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei 112, Taiwan; (C.-H.L.); (S.-J.C.); (F.-S.J.)
| | - Jia-Je Li
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (J.-J.L.); (S.-L.L.); (B.-H.Y.)
| | - Syue-Liang Lin
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (J.-J.L.); (S.-L.L.); (B.-H.Y.)
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Biomedical Engineering Research and Development Center Industrial, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Cheng-Hsiu Lu
- Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei 112, Taiwan; (C.-H.L.); (S.-J.C.); (F.-S.J.)
- Industrial Ph.D Program of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Sain-Jhih Chiu
- Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei 112, Taiwan; (C.-H.L.); (S.-J.C.); (F.-S.J.)
| | - Fong-Shya Jeng
- Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei 112, Taiwan; (C.-H.L.); (S.-J.C.); (F.-S.J.)
| | - Chi-Wei Chang
- National PET and Cyclotron Center (NPCC), Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan;
| | - Bang-Hung Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (J.-J.L.); (S.-L.L.); (B.-H.Y.)
- National PET and Cyclotron Center (NPCC), Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan;
| | - Ming-Cheng Chang
- Institute of Nuclear Energy Research, Atomic Energy Council, Executive Yuan, Taoyuan County 325, Taiwan;
| | - Chien-Chih Ke
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ren-Shyan Liu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei 112, Taiwan; (C.-H.L.); (S.-J.C.); (F.-S.J.)
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (J.-J.L.); (S.-L.L.); (B.-H.Y.)
- Industrial Ph.D Program of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Nuclear Medicine, Cheng Hsin General Hospital, Taipei 112, Taiwan
| |
Collapse
|
47
|
Liu CH, Grodzinski P. Nanotechnology for Cancer Imaging: Advances, Challenges, and Clinical Opportunities. Radiol Imaging Cancer 2021; 3:e200052. [PMID: 34047667 PMCID: PMC8183257 DOI: 10.1148/rycan.2021200052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 02/28/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022]
Abstract
Nanoparticle (NP) imaging applications have the potential to improve cancer diagnostics, therapeutics, and treatment management. In biomedical research and clinical practice, NPs can serve as labels or labeled carriers for monitoring drug delivery or serve as imaging agents for enhanced imaging contrast, as well as providing improved signal sensitivity and specificity for in vivo imaging of molecular and cellular processes. These qualities offer exciting opportunities for NP-based imaging agents to address current limitations in oncologic imaging. Despite substantial advancements in NP design and development, very few NP-based imaging agents have translated into clinics within the past 5 years. This review highlights some promising NP-enabled imaging techniques and their potential to address current clinical cancer imaging limitations. Although most examples provided herein are from the preclinical space, discussed imaging solutions could offer unique in vivo tools to solve biologic questions, improve cancer treatment effectiveness, and inspire clinical translation innovation to improve patient care. Keywords: Molecular Imaging-Cancer, Molecular Imaging-Nanoparticles, Molecular Imaging-Optical Imaging, Metastases, Oncology, Surgery, Treatment Effects.
Collapse
Affiliation(s)
- Christina H. Liu
- From the Cancer Imaging Program, National Cancer Institute, National
Institutes of Health, 9609 Medical Center Dr, Room 4W216, Rockville, MD
20850
| | - Piotr Grodzinski
- From the Cancer Imaging Program, National Cancer Institute, National
Institutes of Health, 9609 Medical Center Dr, Room 4W216, Rockville, MD
20850
| |
Collapse
|
48
|
Zhang C, Chen W, Zhang T, Jiang X, Hu Y. Hybrid nanoparticle composites applied to photodynamic therapy: strategies and applications. J Mater Chem B 2021; 8:4726-4737. [PMID: 32104868 DOI: 10.1039/d0tb00093k] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Photodynamic therapy (PDT), as a robust strategy, has long been applied to cancer treatment owing to the meaningful breakthroughs and unique advantages, including ignorable invasiveness and spatio-temporal selectivity. Numerous PDT agents, especially hybrid nanoparticle composite (hybrid)-based sensitizers consisting of an organic polymer and inorganic nanoparticles (NPs), feature the synergetic pros of the components, which have unlocked the additional potentials of PDT. Although reviews relating to the applications of hybrids to PDT have been previously reported, most of them only focus on the designs of smart hybrids integrating multimodal imaging-guided multiple treatment modalities. Traditional PDT treatment has several limitations, such as inadequate PDT agents accumulating in cancer tissues, inferior PDT effect due to the devastating cancer hypoxia environment, relevant systemic toxicity in non-intelligent stimulation response treatment systems, and serious dependence of PDT on external light sources. Many strategies have been developed for overcoming these limitations, including improvement of cancer-homing ability by introducing active targeting groups, remodeling of the cancer hypoxia environment through oxygen regulators, intratumor release of ROS through activatable molecules, and replacement of laser light by X-rays or self-luminescence. This review aims to summarize the most recent advances in designing hybrids for improving the therapeutic efficacy of PDT.
Collapse
Affiliation(s)
- Chao Zhang
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, China. and Shenzhen Research Institute of Nanjing University, Shenzhen, 518057, China
| | - Weizhi Chen
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210093, China.
| | - Taixing Zhang
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, China.
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210093, China.
| | - Yong Hu
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, China. and Shenzhen Research Institute of Nanjing University, Shenzhen, 518057, China
| |
Collapse
|
49
|
Wang Y, Gou K, Guo X, Ke J, Li S, Li H. Advances in regulating physicochemical properties of mesoporous silica nanocarriers to overcome biological barriers. Acta Biomater 2021; 123:72-92. [PMID: 33454385 DOI: 10.1016/j.actbio.2021.01.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/16/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022]
Abstract
Mesoporous silica nanoparticles (MSNs) with remarkable structural features have been proven to be an excellent platform for the delivery of therapeutic molecules. Biological barriers in various forms (e.g., mucosal barrier, cellular barrier, gastrointestinal barrier, blood-brain barrier, and blood-tumor barrier) present substantial obstacles for MSNs. The physicochemical parameters of MSNs are known to be effective and tunable not only for load and release of therapeutic molecules but also for their biological responsiveness that is beneficial for cells and tissues. This review innovatively provides a description of how and why physicochemical properties (e.g., particle size, morphology, surface charge, hydrophilic-hydrophobic property, and surface modification) of MSNs influence their ability to cross the biological barriers prior to reaching targeted sites. First, the structural and physiological features of biological barriers are outlined. Next, the recent progresses in the critical physicochemical parameters of MSNs are highlighted from physicochemical and biological aspects. Surface modification, as an important strategy for achieving rapid transport, is also reviewed with special attention to the latest findings of bioactive groups and molecular mechanisms. Furthermore, advanced designs of multifunction intelligent MSNs to surmount the blood-tumor barrier and to actively target tumor sites are demonstrated in detail. Lastly, the biodegradability and toxicity of MSNs are evaluated. With perspectives for their potential application and biosafety, the clues in summary might lead to drug delivery with high efficiency and provide useful knowledge for rational design of nanomaterials.
Collapse
|
50
|
Jiménez-Mancilla NP, Aranda-Lara L, Morales-Ávila E, Camacho-López MA, Ocampo-García BE, Torres-García E, Estrada-Guadarrama JA, Santos-Cuevas CL, Isaac-Olivé K. Electron transfer reactions in rhodamine: Potential use in photodynamic therapy. J Photochem Photobiol A Chem 2021. [DOI: 10.1016/j.jphotochem.2021.113131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|