1
|
Shu Z, Guo W, Shang Y, Yang Y, Yang P. Engineering copper (II) in situ reduction and delivery by a novel amphiphilic diacylhydrazone for chemodynamic therapy. Bioorg Chem 2025; 161:108545. [PMID: 40339503 DOI: 10.1016/j.bioorg.2025.108545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/10/2025] [Accepted: 04/27/2025] [Indexed: 05/10/2025]
Abstract
Chemodynamic treatment (CDT) possesses the ability to disturb intracellular redox equilibrium, thus inhibiting tumor development. The copper-induced CDT generally needs the delivery of copper (II) ions to tumor cells and reacting with local GSH, to produce Cu+, and thus generating toxic hydroxyl radicals (·OH) via a Fenton-like reaction. Due to an insufficient amount of endogenous GSH, it remains a great challenge to achieve satisfactory anticancer efficacy and the development of a receptor with the capability of both binding and reducing Cu (II) ions is valuable. In this study, we synthesized a novel amphiphilic diacylhydrazone (compound 1) by a brief chemical reaction. Compound 1 was not only capable of complexation with copper(II) ions but able to reduce the divalent copper ions partially to the monovalent ones. The generated assembly (complex 1) containing mixed Cu (I)/Cu (II) presents a spherical-like nanoparticle, which could easily enter the HCT-116 cells and release both Cu+ and Cu2+ in a mildly acidic environment. The complex 1 therefore inhibited the growth of HCT-116 cells due to the mitochondrial damage initiated by the enhanced Fenton-like effect and the growth inhibition rate of in vivo colon cancer proliferation reached 85.12 %, significantly higher than that of its components of either compound 1 or Cu(ClO4)2, as well as paclitaxel (the positive control). Meanwhile, complex 1 had relatively low toxicity. This work provided a reference for transit Cu (I/II)-mediated precision-targeted CDT therapeutic regimen.
Collapse
Affiliation(s)
- Zhengning Shu
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, China
| | - Wanxin Guo
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, China
| | - Yongxin Shang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, China
| | - Yi Yang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, China
| | - Peng Yang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, China.
| |
Collapse
|
2
|
Ma D, Lu H, Zhang H, Liao A, Peng Q, Yang M. ICG/MnO 2-HFn-mPEG-DSPE-Lip enhances the anticancer activity of ICG phototherapy. Lasers Med Sci 2025; 40:243. [PMID: 40411641 DOI: 10.1007/s10103-025-04494-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 05/11/2025] [Indexed: 05/26/2025]
Abstract
Hypoxia poses a significant challenge to the efficacy of photodynamic therapy (PDT) for cancer treatment. This study aims to design and synthesize PEGylated liposomes encapsulating MnO₂, indocyanine green (ICG), and H-chain ferritin (HFn) to potentially address hypoxia and enhance the therapeutic outcomes of PDT. PEGylated liposomes (ICG/MnO₂-HFn-mPEG-DSPE-Lip) were constructed with a rod-like structure, incorporating MnO₂ as a hypoxia-modulating agent and ICG as a photosensitizer. The drug loading capacity, stability and safety of liposomes were characterized. Singlet oxygen quantum yield (ΦΔ) was measured under simulated tumor microenvironment conditions. In vitro phototoxicity was evaluated using A549 human lung adenocarcinoma cells. Liposomes have a high drug loading capacity, good biocompatibility and good long-term stability. Under tumor-simulated conditions, ΦΔ was significantly improved, increasing from 0.210 for free ICG to 0.507. The liposomes demonstrated remarkable phototoxic effects on A549 cells (90.5% cell death under combined PDT/PTT vs. 15% for free ICG). This nanoplatform proposes a novel strategy to overcome hypoxia-induced PDT resistance, and the enhanced efficacy may be attributed to the increased oxygen supply mediated by MnO2 carried by HFn. These findings provide important insights for the development of next-generation therapeutic systems targeting tumor hypoxia.
Collapse
Affiliation(s)
- Dan Ma
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Huixiang Lu
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Hai Zhang
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Anru Liao
- College of Pharmacy, Guizhou University, Guiyang, China
| | - Qianrong Peng
- China Tobacco Guizhou Industrial Co., Ltd., Guiyang, China
| | - Min Yang
- College of Pharmacy, Guizhou University, Guiyang, China.
| |
Collapse
|
3
|
Eladarousy O, Zibara Z, Hussari K, Al Helou N, Nicolas R. Optimizing plasmonic photothermal therapy a numerical study on tissue specific thermal and optical properties for cancer treatment. Sci Rep 2025; 15:17897. [PMID: 40410282 PMCID: PMC12102199 DOI: 10.1038/s41598-025-95798-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/24/2025] [Indexed: 05/25/2025] Open
Abstract
Effective cancer treatment demands precision to target malignant tissues while sparing healthy ones. This study investigates numerically the role of plasmonic photothermal therapy (PPTT) as a minimally invasive approach to achieve this goal, focusing on the interplay between nanoparticle (NP) size, resonance wavelength, and the thermal properties of biological tissues. Tailoring NP characteristics to align with tissue-specific optical and thermal properties enables precise thermal delivery, enhancing therapeutic efficacy while minimizing collateral damage. Resonance wavelength shifts with NP size, while tissue parameters like thermal conductivity and refractive index further influence temperature profiles. These findings highlight the importance of optimizing PPTT protocols through computational modeling and experimental validation. This approach advances personalized cancer therapies, offering safer and more effective treatment outcomes.
Collapse
Affiliation(s)
- Omar Eladarousy
- Department of Physical Sciences, Lebanese American University, Beirut, Lebanon
| | - Zahraa Zibara
- Department of Biological Sciences, Lebanese American University, Beirut, Lebanon
| | - Khaled Hussari
- Department of Biological Sciences, Lebanese American University, Beirut, Lebanon
| | - Nissrine Al Helou
- Department of Physical Sciences, Lebanese American University, Beirut, Lebanon
| | - Rana Nicolas
- Department of Physical Sciences, Lebanese American University, Beirut, Lebanon.
| |
Collapse
|
4
|
Sahu A, Ingle J, Panigrahi R, Basu S. Small Molecule-Mediated Photothermal Therapy Induces Apoptosis in Cancer Cells. ChemMedChem 2025:e2500151. [PMID: 40346638 DOI: 10.1002/cmdc.202500151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/11/2025]
Abstract
Cancer remains as one of the most life-threatening diseases in the whole world. Most of the therapeutic strategies to eradicate cancer are highly invasive, leading to severe injury and trauma to the patients. In recent times, phototherapy has emerged as one of the noninvasive therapeutic strategies for cancer treatment. However, development of novel small-molecule photothermal agents remains a major challenge. To address this, herein, a small molecule library having aromatic substituted-3-methoxy-pyrrole and 2-(3-cyano-4,5,5-trimethylfuran-2(5 H)-ylidene) malononitrile in a concise synthetic strategy is designed and synthesized. One of the library members (7H) self-assembles into spherical-like nanoparticles having <100 nm size in water and is found to exhibit remarkable increase in temperature under 740 nm near-infrared (NIR) light. Interestingly, compound 7H homes into the lysosomal compartments and the lipid droplets in the HCT-116 colon cancer cells within 3 h and induces photothermal effect followed by generation of reactive oxygen species while irradiating under 740 nm NIR light for 10 min. Moreover, 7H triggers programmed cell death (apoptosis) to induce remarkable HCT-116 cell killing. This small molecule-mediated photothermal effect shows potential to be an interesting tool for the next-generation noninvasive cancer phototherapy.
Collapse
Affiliation(s)
- Asima Sahu
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj Gandhinagar, 382355, India
| | - Jaypalsing Ingle
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj Gandhinagar, 382355, India
| | - Reha Panigrahi
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj Gandhinagar, 382355, India
| | - Sudipta Basu
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj Gandhinagar, 382355, India
| |
Collapse
|
5
|
Shao Y, Song J, Hao C, Lv F, Hou H, Fan X, Song F. A simple co-assembly strategy to control the dimensions of nanoparticles for enhanced synergistic therapy. J Colloid Interface Sci 2025; 685:1008-1017. [PMID: 39879780 DOI: 10.1016/j.jcis.2025.01.216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Despite phthalocyanine has excellent photodynamic and photothermal effects as a photosensitizer and photothermal agent, hydrophobicity and aggregation limits its biological application. In this paper, phthalocyanine-cyanine co-assembled nanoparticles were designed to modulate the dimensions and morphology by introducing water-soluble cyanine. The cyanine had the ability to transform the nanomaterials from microrods to nanospheres, thus successfully constructing photoactivated nanomedicines. Their appropriate size effect and improved water solubility conferred the nanoparticles with extended blood circulation time and tumor accumulation capacity. Meanwhile, the fluorescence effect of cyanine enabled the nanoparticles to have the ability of fluorescence imaging. The nanoparticles achieved enhanced PDT/PTT synergistic effect under single laser induction, especially the generation of type I photodynamics.
Collapse
Affiliation(s)
- Yutong Shao
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao 266237 PR China
| | - Jitao Song
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao 266237 PR China.
| | - Caiqin Hao
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao 266237 PR China
| | - Fangyuan Lv
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao 266237 PR China
| | - Haoran Hou
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024 PR China
| | - Xinping Fan
- Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), PR China.
| | - Fengling Song
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao 266237 PR China.
| |
Collapse
|
6
|
Wang Z, Zhu B, Nie W, Zhang L, Xiao N, Zhang Q, Wu Z, Shi C, Zhu W, Liu Q, Zhu D, Bryce MR, Ren L, Tang BZ. Endogenous Near-Infrared Chemiluminescence: Imaging-Guided Non-Invasive Thrombolysis and Anti-Inflammation Based on a Heteronuclear Transition Metal Complex. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2501257. [PMID: 40135831 PMCID: PMC12097061 DOI: 10.1002/advs.202501257] [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] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/28/2025] [Indexed: 03/27/2025]
Abstract
Conventional therapy to treat thrombi (blood clots) has significant limitations: i) inflammation; ii) bleeding side effects; iii) re-embolisation, and iv) in situ thrombi that are not visible. Here it is reported that Cu2Ir nanoparticles (NPs) with a Cu-coordinated tetraphenylporphyrin (TPP) core and cyclometalated Ir(C^N)2(N^N) substituents integrate long-lived near-infrared (NIR) chemiluminescence (CL) imaging, photothermal therapy (PTT) and photodynamic therapy (PDT) for thrombolysis, with antioxidant and anti-inflammatory properties. Based on density functional theory calculations the chemiluminescent reaction site between TPP and peroxynitrite (ONOO-) is confirmed for the first time. The presence of the transition metal significantly improves the chemiluminescent properties of TPP. Upon specific activation by ONOO-, Cu2Ir NPs exhibited more than 30-fold NIR CL intensity than TPP NPs, and the luminescence lasted for 60 min allowing for precise and long-lasting dynamic tracking of thrombi. Cu2Ir NPs achieved non-invasive safe thrombolytic therapy triggered by NIR irradiation at the signaling site. 72.3% blood reperfusion is obtained for nearly complete restoration of blood flow, and re-embolism is prevented in a mouse carotid artery model. Furthermore, Cu2Ir NPs scavenged excess reactive oxygen/nitrogen species (RONS) and reduced inflammatory factors. Cu2Ir NPs hold promise as a single-molecule strategy for diagnosing and treating diseases associated with thrombosis.
Collapse
Affiliation(s)
- Ziwei Wang
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin ProvinceDepartment of ChemistryNortheast Normal University5268 Renmin StreetChangchunJilin130024P. R. China
| | - Bo Zhu
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin ProvinceDepartment of ChemistryNortheast Normal University5268 Renmin StreetChangchunJilin130024P. R. China
| | - Wenxin Nie
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin ProvinceDepartment of ChemistryNortheast Normal University5268 Renmin StreetChangchunJilin130024P. R. China
| | - Liping Zhang
- Department of NeurologyInst Translat MedThe First Affiliated Hospital of Shenzhen UniversityShenzhen Second People's HospitalShenzhen518035P. R. China
| | - Nan Xiao
- Department of NeurologyInst Translat MedThe First Affiliated Hospital of Shenzhen UniversityShenzhen Second People's HospitalShenzhen518035P. R. China
| | - Qiaohua Zhang
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin ProvinceDepartment of ChemistryNortheast Normal University5268 Renmin StreetChangchunJilin130024P. R. China
| | - Zihan Wu
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin ProvinceDepartment of ChemistryNortheast Normal University5268 Renmin StreetChangchunJilin130024P. R. China
| | - Chunguang Shi
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin ProvinceDepartment of ChemistryNortheast Normal University5268 Renmin StreetChangchunJilin130024P. R. China
| | - Weijin Zhu
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin ProvinceDepartment of ChemistryNortheast Normal University5268 Renmin StreetChangchunJilin130024P. R. China
| | - Qianwen Liu
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin ProvinceDepartment of ChemistryNortheast Normal University5268 Renmin StreetChangchunJilin130024P. R. China
| | - Dongxia Zhu
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin ProvinceDepartment of ChemistryNortheast Normal University5268 Renmin StreetChangchunJilin130024P. R. China
| | | | - Lijie Ren
- Department of NeurologyInst Translat MedThe First Affiliated Hospital of Shenzhen UniversityShenzhen Second People's HospitalShenzhen518035P. R. China
| | - Ben Zhong Tang
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionState Key Laboratory of Molecular NeuroscienceDivision of Life ScienceMing Wai Lau Centre for Reparative MedicineKarolinska Instituteand Guangdong‐Hong Kong‐Macau Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyKowloonHong Kong999077P. R. China
- School of Science and EngineeringShenzhen Institute of Aggregate Science and TechnologyThe Chinese University of Hong KongShenzhenGuangdong518172P. R. China
| |
Collapse
|
7
|
Cesca BA, Pellicer San Martin K, Caverzan MD, Oliveda PM, Ibarra LE. State-of-the-art photodynamic therapy for malignant gliomas: innovations in photosensitizers and combined therapeutic approaches. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002303. [PMID: 40177536 PMCID: PMC11964779 DOI: 10.37349/etat.2025.1002303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/11/2025] [Indexed: 04/05/2025] Open
Abstract
Glioblastoma (GBM), the most aggressive and lethal primary brain tumor, poses a significant therapeutic challenge due to its highly invasive nature and resistance to conventional therapies, including surgery, chemotherapy, and radiotherapy. Despite advances in standard treatments, patient survival remains limited, requiring the exploration of innovative strategies. Photodynamic therapy (PDT) has emerged as a promising approach, leveraging light-sensitive photosensitizers (PSs), molecular oxygen, and specific light wavelengths to generate reactive oxygen species (ROS) that selectively induce tumor cell death. Originally developed for skin cancer, PDT has evolved to target more complex malignancies, including GBM. The refinement of second- and third-generation PS, coupled with advancements in nanotechnology, has significantly improved PDT's selectivity, bioavailability, and therapeutic efficacy. Moreover, the combination of PDT with chemotherapy, targeted therapy, and immunotherapy, among other therapeutic modalities, has shown potential in enhancing therapeutic outcomes. This review provides a comprehensive analysis of the preclinical and clinical applications of PDT in GBM, detailing its mechanisms of action, the evolution of PS, and novel combinatory strategies that optimize treatment efficacy. However, several challenges remain, including overcoming GBM-associated hypoxia, enhancing PS delivery across the blood-brain barrier, and mitigating tumor resistance mechanisms. The integration of PDT with molecular and genetic insight, alongside cutting-edge nanotechnology-based delivery systems, may revolutionize GBM treatment, offering new prospects for improved patient survival and quality of life.
Collapse
Affiliation(s)
- Bruno A. Cesca
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Río Cuarto (UNRC), Rio Cuarto X5800BIA, Argentina
| | - Kali Pellicer San Martin
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Río Cuarto (UNRC), Rio Cuarto X5800BIA, Argentina
| | - Matías D. Caverzan
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Río Cuarto X5800BIA, Argentina
- Departamento de Patología Animal, Facultad de Agronomía y Veterinaria, Universidad Nacional de Río Cuarto (UNRC), Rio Cuarto X5800BIA, Argentina
| | - Paula M. Oliveda
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Río Cuarto (UNRC), Rio Cuarto X5800BIA, Argentina
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rio Cuarto X5800BIA, Argentina
| | - Luis E. Ibarra
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Río Cuarto (UNRC), Rio Cuarto X5800BIA, Argentina
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rio Cuarto X5800BIA, Argentina
| |
Collapse
|
8
|
Shi H, Marchi RC, Sadler PJ. Advances in the Design of Photoactivatable Metallodrugs: Excited State Metallomics. Angew Chem Int Ed Engl 2025; 64:e202423335. [PMID: 39806815 DOI: 10.1002/anie.202423335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Indexed: 01/16/2025]
Abstract
Photoactivatable metal complexes offer the prospect of novel drugs with low side effects and new mechanisms of action to combat resistance to current therapy. We highlight recent progress in the design of platinum, ruthenium, iridium, gold and other transition metal complexes, especially for applications as anticancer and anti-infective agents. In particular, understanding excited state chemistry related to identification of the bioactive species (excited state metallomics/pharmacophores) is important. Photoactivatable metallodrugs are classified here as photocatalysts, photorelease agents and ligand-activated agents. Their activation wavelengths, cellular mechanisms of action, experimental and theoretical metallomics of excited states and photoproducts are discussed to explore new strategies for the design and investigation of photoactivatable metallodrugs. These photoactivatable metallodrugs have potential in clinical applications of Photodynamic Therapy (PDT), Photoactivated Chemotherapy (PACT) and Photothermal Therapy (PTT).
Collapse
Affiliation(s)
- Huayun Shi
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, China
| | - Rafael C Marchi
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - Peter J Sadler
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
9
|
Goetzfried SK, Hakkennes MLA, Busemann A, Bonnet S. Toward the Treatment of Glioblastoma Tumors Using Photoactivated Chemotherapy: In Vitro Evaluation of Efficacy and Safety. ACS Pharmacol Transl Sci 2025; 8:484-498. [PMID: 39974641 PMCID: PMC11833736 DOI: 10.1021/acsptsci.4c00600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/21/2025]
Abstract
Glioblastoma multiforme (GBM) is highly aggressive, necessitating new therapies. Photoactivated chemotherapy (PACT) offers a promising approach by activating prodrugs with visible light at the tumor site. This study evaluated the anticancer activity of ruthenium-based PACT compounds in U-87MG glioblastoma cells and their safety in SH-SY5Y neuron-like cells. The compound [3](PF6)2 showed promising light-activated anticancer effects in U-87MG cells, while [1](PF6)2 was inactive, and [2](PF6)2 was nonactivated. Interestingly, in SH-SY5Y cells, light-activated [3](PF6)2 increased cell proliferation, similar to donepezil, without causing cell death. Increased Ca2+ uptake was observed, possibly via interaction with the AMPA receptor, as suggested by docking studies. These findings suggest ruthenium-based PACT compounds may serve as potential treatments for GBM, effectively attacking cancer cells while preserving healthy neuronal cells.
Collapse
Affiliation(s)
| | - Matthijs L. A. Hakkennes
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| | - Anja Busemann
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| |
Collapse
|
10
|
Dutta J, Varshini A, Padaga SG, Bera A, Sarkar T, Biswas S, Hussain A. Red and NIR light-triggered enhancement of anticancer and antibacterial activities of dinuclear Co(II)-catecholate complexes. Dalton Trans 2025; 54:3027-3038. [PMID: 39812808 DOI: 10.1039/d4dt03153a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Photoactive complexes of bioessential 3d metals, activable within the phototherapeutic window (650-900 nm), have gained widespread interest due to their therapeutic potential. Herein, we report the synthesis, characterization, and light-enhanced anticancer and antibacterial properties of four new dinuclear Co(II) complexes: [Co(phen)(cat)]2 (Co-1), [Co(dppz)(cat)]2 (Co-2), [Co(phen)(esc)]2 (Co-3), and [Co(dppz)(esc)]2 (Co-4). In these complexes, phen (1,10-phenanthroline) and dppz (dipyrido[3,2-a:2',3'-c]phenazine) act as neutral N,N-donor ligands, while cat2- and esc2- serve as O,O-donor catecholate ligands derived from catechol (1,2-dihydroxybenzene) and esculetin (6,7-dihydroxy coumarin). Their high-spin paramagnetic nature and dimeric identity in solution were confirmed by magnetic susceptibility, UV-visible, emission, and mass spectral data. Co-1-Co-4 exhibited an absorption band within the 600-850 nm range, originating from a charge transfer transition. The electrically neutral complexes demonstrated sufficient solution stability both in the dark and under irradiated conditions. The dppz complexes Co-2 and Co-4 exhibited notable toxicity towards A549 lung carcinoma cells, with potency increasing significantly under brief (5 min) exposure to 660 nm (red) and 808 nm (NIR) laser light (IC50 ∼ 8.9 to 14.9 μM). Notably, their toxicity towards normal NIH-3T3 fibroblast cells was minimal. Cellular assays highlighted that the induced cell death followed an apoptotic pathway, primarily due to mitochondrial damage. Co-2 and Co-4 also demonstrated significant antibacterial potency against Gram-(+) S. aureus and Gram-(-) P. aeruginosa, with effectiveness significantly enhanced upon 808 nm laser irradiation (MIC ∼ 15-142 μM). The increase in the anticancer and antibacterial efficacies was attributed to the generation of cytotoxic singlet oxygen (1O2) species upon red/NIR light exposure. Notably, 808 nm NIR irradiation produced more pronounced effects compared to 660 nm. This study is the first to report on cobalt complexes exhibiting red and NIR light-triggered enhancement of antibacterial and anticancer activities, illuminating the path for the development of long-wavelength absorbing cobalt complexes with enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Jyotirmoy Dutta
- Department of Chemistry, Handique Girls' College, Guwahati 781001, Assam, India.
| | - Are Varshini
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India.
| | - Sri Ganga Padaga
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India.
| | - Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bengaluru, 560012, Karnataka, India
| | - Tukki Sarkar
- Department of Fluoro-Agrochemicals, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India.
| | - Swati Biswas
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India.
| | - Akhtar Hussain
- Department of Chemistry, Handique Girls' College, Guwahati 781001, Assam, India.
| |
Collapse
|
11
|
Liu X, Yuan H. Responsive nanomaterials in biomedicine, patent path and prospect analysis. Front Bioeng Biotechnol 2025; 13:1539991. [PMID: 39968009 PMCID: PMC11832473 DOI: 10.3389/fbioe.2025.1539991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
In recent years, responsive nanomaterials have demonstrated tremendous potential in biomedical applications due to their unique advantages in precise drug delivery and controlled release. For complex diseases such as cancer, chronic inflammation, and genetic disorders, traditional treatment methods are often limited by insufficient targeting and significant side effects. Responsive nanotechnology, by sensing specific internal or external stimuli, has significantly enhanced the precision and efficiency of treatments. This study systematically summarizes the technological trajectory and emerging research directions of responsive nanomaterials through global patent and literature data, employing main path analysis, derivative path analysis, and keyword co-occurrence analysis. The results reveal the evolution of this field, from the optimization of early single-stimulus-responsive nano delivery systems to the rise of theranostics integration, followed by advancements in multi-stimuli-responsive synergistic therapies, and finally, the innovation in biomimetic material design. Each developmental phase has increasingly focused on adapting to complex biological environments, achieving superior targeting performance, and enhancing therapeutic efficacy. Keyword co-occurrence analysis highlights key research hotspots, including biomimetic design, multimodal synergistic therapies, and emerging response mechanisms. In the future, responsive nanomaterials are expected to play a pivotal role in personalized medicine, multifunctional carrier design, and complex disease management, providing novel insights and technological support for precision medicine.
Collapse
Affiliation(s)
| | - Hongmei Yuan
- School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
12
|
Xu X, Sun J, Zhao J, Yu F, Xu Y, Zhang C, Li J. Cell membrane-camouflaged nanoarchitectonics of photosensitizer nanoparticles for enhanced phototherapy in surgery. J Colloid Interface Sci 2025; 679:726-736. [PMID: 39393150 DOI: 10.1016/j.jcis.2024.09.225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/13/2024]
Abstract
Surgical risk and wound area can be reduced by diminishing tumor volume before surgery. The chemotherapy and radiotherapy currently used that can reduce the tumor volume generally cause severe systemic side effects. Phototherapy has recently emerged as an effective treatment modality for superficial cancers. However, phototherapy is limited by the low utilization of photosensitizer, the tumor hypoxia, and the low photothermal conversion efficiency. Herein, we report the cancer membrane biomimetic nanoparticles assembled by Chlorin e6 (Ce6) and chlorambucil (CRB). Ce6@CRB nanoparticles (CCNPs) show excellent photothermal conversion efficiency, which is 2 times higher than free Ce6. Meanwhile, CCNPs can produce singlet oxygen stably compared to free Ce6 thereby reducing the dependence on oxygen. Furthermore, the coating of 4 T1 cancer membrane on the surface of CCNPs endows them with the ability of homologous targeting, not only improving the utilization of Ce6, but also effectively activating the immune system in vivo when combined photodynamic therapy (PDT) and photothermal therapy (PTT). Intriguingly, surgical resection is performed after phototherapy in this treatment regimen, which can effectively reduce the wound area. Together, this work provided a feasible and creative method for tumor clinical therapy for its patient-centric and humanitarian focus.
Collapse
Affiliation(s)
- Xia Xu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Jiachen Sun
- Department of Dermatology, Peking University Third Hospital, Beijing 100191, China
| | - Jie Zhao
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Fanchen Yu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Xu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunlei Zhang
- Department of Dermatology, Peking University Third Hospital, Beijing 100191, China.
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
13
|
Frumento D, Ţălu Ș. Light-based technologies in immunotherapy: advances, mechanisms and applications. Immunotherapy 2025; 17:123-131. [PMID: 40032620 PMCID: PMC11901425 DOI: 10.1080/1750743x.2025.2470111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025] Open
Abstract
Light-based immunotherapy uses specific wavelengths of light to activate or modulate immune responses. It primarily employs two mechanisms: direct activation of immune cells and indirect modulation of the tumor microenvironment (TME). Several light-based technologies are under investigation or clinical use in immunotherapy, including photodynamic immunotherapy (PDIT) and photothermal therapy (PTT). Optogenetic tools have the potential to precisely control T-cell receptor activation, cytokine release, or the activity of other immune effector cells. Light-based technologies present innovative opportunities within the realm of immunotherapy. The ability to precisely regulate immune cell activation via optogenetics, alongside the improved targeting of cancer cells through photoimmunotherapy, signifies a transformative shift in our strategies for immune modulation. Although many of these technologies remain in the experimental stage for various applications, initial findings are encouraging, especially concerning cancer treatment and immune modulation. Continued research and clinical trials are essential to fully harness the capabilities of light technology in the context of immune cell therapy.
Collapse
Affiliation(s)
| | - Ștefan Ţălu
- The Directorate of Research, Development and Innovation Management (DMCDI), The Technical University of Cluj-Napoca, Cluj-Napoca, Romania
| |
Collapse
|
14
|
Gao Q, Xu Y, Galluzzi M, Xing Q, Geng J. Enhanced Cancer Cell Specificity Through Combined Blue Light Therapy and Starvation Strategies. Adv Biol (Weinh) 2025; 9:e2400264. [PMID: 39617743 DOI: 10.1002/adbi.202400264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/22/2024] [Indexed: 02/18/2025]
Abstract
In this study, the effectiveness of combining short-term starvation (STS or fasting) is investigated with blue light illumination therapy in delaying the progression of various types of cancer, including osteosarcoma, cervical, breast, liver carcinoma, and melanoma cancer in animal models. Moreover, the comparative analysis between cancerous (including HeLa, 143B, MDA-MB-231, and HepG2) and normal cell lines (including NCM460, HEKa, and L-O2), highlights the selectivity of the treatment's cytotoxic effects, favoring cancer cells while largely sparing normal cells. In HeLa cancer cells, treatment with the STS and blue light illumination combination resulted in increased phosphorylation of JNK and p38, which led to the activation of downstream signalling substrates, such as p53 and H2AX. This activation induced mitochondrial and nuclear damage, ultimately leading to tumor cell death. The combination treatment also caused metabolic disorders in tumor cells, which interfered with biomolecule availability and selectively induced lethal effects in tumor cells. Therefore, the combination treatment can be an effective strategy for eliminating cancer.
Collapse
Affiliation(s)
- Quan Gao
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Youwei Xu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Massimiliano Galluzzi
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qi Xing
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jin Geng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
15
|
Shi H, Mohan M, Singh K, Sadler PJ. An optical coherence tomography study of a photoactive Pt(iv) prodrug in oesophageal tissue. RSC Adv 2025; 15:3168-3171. [PMID: 39896429 PMCID: PMC11783044 DOI: 10.1039/d4ra08693g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/10/2025] [Indexed: 02/04/2025] Open
Abstract
Photoactive diazido Pt(iv) complexes display in vivo anticancer efficacy towards oesophageal tumours, a worldwide common cancer. Here we explore the use of optical coherence tomography (OCT) as a new method for detecting tissue penetration and damage produced by the photoactivatable anticancer complex trans,trans,trans-[Pt(pyridine)2(N3)2(OH)2] (FM190). Dehydration of the sample and a change in refractive index were observed for swine oesophageal tissue treated with FM190 and blue laser light (445 nm) using an OCT system. In contrast, tissues treated with FM190 or laser light alone showed no apparent damage.
Collapse
Affiliation(s)
- Huayun Shi
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University Xiamen 361102 China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University Xiamen 361102 China
| | - Muktesh Mohan
- Max-Planck-Institut für die Physik des Lichts Staudtstraße 2 91058 Erlangen Germany
- Max-Planck-Zentrum für Physik und Medizin Kussmaulallee 2 91054 Erlangen Germany
| | - Kanwarpal Singh
- Max-Planck-Institut für die Physik des Lichts Staudtstraße 2 91058 Erlangen Germany
- Max-Planck-Zentrum für Physik und Medizin Kussmaulallee 2 91054 Erlangen Germany
- Department of Electrical and Computer Engineering, McMaster University 1280 Main Street West Hamilton ON L8S 4K1 Canada
| | - Peter J Sadler
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| |
Collapse
|
16
|
Baek M, Lee SM, Kim D, Lee J. Breakthroughs in deep tumour penetrating nano-phototheranostics for tumour ablation. Clin Transl Med 2025; 15:e70188. [PMID: 39799417 PMCID: PMC11726636 DOI: 10.1002/ctm2.70188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Affiliation(s)
- Min‐Jun Baek
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Sang Min Lee
- College of Pharmacy and Research Institute of Pharmaceutical SciencesSeoul National UniversitySeoulRepublic of Korea
| | - Dae‐Duk Kim
- College of Pharmacy and Research Institute of Pharmaceutical SciencesSeoul National UniversitySeoulRepublic of Korea
| | - Jae‐Young Lee
- College of Pharmacy and Research Institute of Pharmaceutical SciencesSeoul National UniversitySeoulRepublic of Korea
| |
Collapse
|
17
|
Parihar A, Gaur K, Sarbadhikary P. Advanced 2D Nanomaterials for Phototheranostics of Breast Cancer: A Paradigm Shift. Adv Biol (Weinh) 2025; 9:e2400441. [PMID: 39543015 DOI: 10.1002/adbi.202400441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/29/2024] [Indexed: 11/17/2024]
Abstract
Breast cancer is the leading cause of women's deaths and associated comorbidities. The advanced and targeted strategies against breast cancer have gained considerable attention due to their potential enhanced therapeutic efficacy over conventional therapies. In this context, phototherapies like photodynamic therapy (PDT) and photothermal therapy (PTT) have shown promise as an effective and alternative strategy due to reduced side effects, noninvasiveness, and spatiotemporal specificity. With the advent of nanotechnology, several types of nanomaterials that have shown excellent prospects in increasing the efficacy of photo therapies have been exploited in cancer treatment. In recent years, 2D nanomaterials have stood out promising because of their unique ultrathin planar structure, chemical, physical, tunable characteristics, and corresponding remarkable physiochemical/biological properties. In this review, the potential and the current status of several types of 2D nanomaterials such as graphene-based nanomaterials, Mxenes, Black phosphorous, and Transition Metal Dichalcogenides for photo/thermo and combination-based imaging and therapy of breast cancer have been discussed. The current challenges and prospects in terms of translational potential in future clinical oncology are highlighted.
Collapse
Affiliation(s)
- Arpana Parihar
- Department of Translational Medicine, All India Institute of Medical Sciences Bhopal, Saket Nagar, Bhopal, Madhya Pradesh, 462020, India
| | - Kritika Gaur
- Central Sheep and wool research institute, ICAR- Indian Council of Agricultural Research, Avikanagr, Malpura, Rajasthan, 304501, India
| | - Paromita Sarbadhikary
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg, 2028, South Africa
| |
Collapse
|
18
|
Li R, Qian J, Zhu X, Tao T, Zhou X. Nanomolecular machines: Pioneering precision medicine for neoplastic diseases through advanced diagnosis and treatment. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167486. [PMID: 39218275 DOI: 10.1016/j.bbadis.2024.167486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Tumors pose a major threat to human health, accounting for nearly one-sixth of global deaths annually. The primary treatments include surgery, radiotherapy, chemotherapy, and immunotherapy, each associated with significant side effects. This has driven the search for new therapies with fewer side effects and greater specificity. Nanotechnology has emerged as a promising field in this regard, particularly nanomolecular machines at the nanoscale. Nanomolecular machines are typically constructed from biological macromolecules like proteins, DNA, and RNA. These machines can be programmed to perform specialized tasks with precise instructions. Recent research highlights their potential in tumor diagnostics-identifying susceptibility genes, detecting viruses, and pinpointing tumor markers. Nanomolecular machines also offer advancements in tumor therapy. They can reduce traditional treatment side effects by delivering chemotherapy drugs and enhancing immunotherapy, and they support innovative treatments like sonodynamic and phototherapy. Additionally, they can starve tumors by blocking blood vessels, and eliminate tumors by disrupting cell membranes or lysosomes. This review categorizes and explains the latest achievements in molecular machine research, explores their models, and practical clinical uses in tumor diagnosis and treatment. It aims to broaden the research perspective and accelerate the clinical adoption of these technologies.
Collapse
Affiliation(s)
- Ruming Li
- Department of Immunology, School of Medicine, Nantong University, Nantong, China; The Second Affiliated Hospital, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Jialu Qian
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Xiao Zhu
- The Second Affiliated Hospital, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China.
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China.
| |
Collapse
|
19
|
Zhang J, Liu Z, Zhang Z, Yang H, Wang H, Yang Z, Xu Y, Li S, Yang D. Recent Advances in Silica-Based Nanomaterials for Enhanced Tumor Imaging and Therapy. ACS APPLIED BIO MATERIALS 2024; 7:7133-7169. [PMID: 39495482 DOI: 10.1021/acsabm.4c01318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Cancer remains a formidable challenge, inflicting profound physical, psychological, and financial burdens on patients. In this context, silica-based nanomaterials have garnered significant attention for their potential in tumor imaging and therapy owing to their exceptional properties, such as biocompatibility, customizable porosity, and versatile functionalization capabilities. This review meticulously examines the latest advancements in the application of silica-based nanomaterials for tumor imaging and therapy. It underscores their potential in enhancing various cancer imaging modalities, including fluorescence imaging, magnetic resonance imaging, computed tomography, positron emission tomography, ultrasound imaging, and multimodal imaging approaches. Moreover, the review delves into their therapeutic efficacy in chemotherapy, radiotherapy, phototherapy, immunotherapy, gas therapy, sonodynamic therapy, chemodynamic therapy, starvation therapy, and gene therapy. Critical evaluations of the biosafety profiles and degradation pathways of these nanomaterials within biological environments are also presented. By discussing the current challenges and prospects, this review aims to provide a nuanced perspective on the clinical translation of silica-based nanomaterials, thereby highlighting their promise in revolutionizing cancer diagnostics, enabling real-time monitoring of therapeutic responses, and advancing personalized medicine.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zilu Liu
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zhijing Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Hui Yang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Hui Wang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zhenlu Yang
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, China
| | - Yunjian Xu
- School of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an 271000, China
- Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Shengke Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| |
Collapse
|
20
|
Liu A, Huang Z, Du X, Duvva N, Du Y, Teng Z, Liao Z, Liu C, Tian H, Huo S. Biodegradable Ruthenium-Rhenium Complexes Containing Nanoamplifiers: Triggering ROS-Induced CO Release for Synergistic Cancer Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403795. [PMID: 38995228 PMCID: PMC11425273 DOI: 10.1002/advs.202403795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 07/03/2024] [Indexed: 07/13/2024]
Abstract
The constrained effectiveness of photodynamic therapy (PDT) has impeded its widespread use in clinical practice. Urgent efforts are needed to address the shortcomings faced in photodynamic therapy, such as photosensitizer toxicity, short half-life, and limited action range of reactive oxygen species (ROS). In this study, a biodegradable copolymer nanoamplifier is reported that contains ruthenium complex (Ru-complex) as photosensitizer (PS) and rhenium complex (Re-complex) as carbon monoxide (CO)-release molecule (CORM). The well-designed nanoamplifier brings PS and CORM into close spatial proximity, significantly promotes the utilization of light-stimulated reactive oxygen species (ROS), and cascaded amplifying CO release, thus enabling an enhanced synergistic effect of PDT and gas therapy for cancer treatment. Moreover, owing to its intrinsic photodegradable nature, the nanoamplifier exhibits good tumor accumulation and penetration ability, and excellent biocompatibility in vivo. These findings suggest that the biodegradable cascaded nanoamplifiers pave the way for a synergistic and clinically viable integration of photodynamic and gas therapy.
Collapse
Affiliation(s)
- Aijie Liu
- State Key Laboratory of Cellular Stress BiologyFujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenFujian361102China
- Shenzhen Research Institute of Xiamen UniversityShenzhenGuangdong518057China
| | - Zhenkun Huang
- State Key Laboratory of Cellular Stress BiologyFujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenFujian361102China
| | - Xiangfu Du
- State Key Laboratory of Cellular Stress BiologyFujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenFujian361102China
| | - Naresh Duvva
- Department of Chemistry‐Ångström LaboratoryBox 523 Uppsala UniversityUppsalaSE‐75120Sweden
| | - Yuting Du
- State Key Laboratory of Cellular Stress BiologyFujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenFujian361102China
| | - Zihao Teng
- State Key Laboratory of Cellular Stress BiologyFujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenFujian361102China
| | - Zhihuan Liao
- State Key Laboratory of Cellular Stress BiologyFujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenFujian361102China
| | - Chen Liu
- State Key Laboratory of Cellular Stress BiologyFujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenFujian361102China
| | - Haining Tian
- Department of Chemistry‐Ångström LaboratoryBox 523 Uppsala UniversityUppsalaSE‐75120Sweden
| | - Shuaidong Huo
- State Key Laboratory of Cellular Stress BiologyFujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenFujian361102China
| |
Collapse
|
21
|
Navyatha B, Nara S. The effects of conjugating anti-MUC1 aptamers on gold nanobipyramids and nanostars for photothermal cancer ablation. Nanomedicine (Lond) 2024; 19:1957-1975. [PMID: 39136402 PMCID: PMC11485751 DOI: 10.1080/17435889.2024.2384351] [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: 03/09/2024] [Accepted: 07/22/2024] [Indexed: 10/09/2024] Open
Abstract
Aim: To ascertain the impact of shape and surface modification of anisotropic nanoparticles on the toxicity and photothermal efficiency toward cancerous cell lines.Methods: Gold nanobipyramids and nanostars surface modified with MUC1 aptamer were used in the current study to explore the toxicity and photothermal efficiency on MCF7 breast cancer cell lines via MTT assay.Results: Surface functionalization with MUC1 aptamer showed significant reduction in % cytotoxicity and increase in % specific internalization of nanostructures into MCF7 cell lines. Further, the photothermal studies accomplished at IC50 concentration for 6 h of treatment and laser exposure for 15 min reported that aptamer-conjugated nanobipyramids were more effective and specific toward MCF7 cell lines than aptamer-conjugated nanostars.Conclusion: This work establishes a platform for the development of tailored photoablation based gold nanostructures for in vivo studies.
Collapse
Affiliation(s)
- Bankuru Navyatha
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, U.P., 211004, India
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Guntur, 522302, Andhra Pradesh,India
| | - Seema Nara
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, U.P., 211004, India
| |
Collapse
|
22
|
Ermakova A. Fluorescent Nanodiamonds for High-Resolution Thermometry in Biology. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1318. [PMID: 39120422 PMCID: PMC11313720 DOI: 10.3390/nano14151318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Optically active color centers in diamond and nanodiamonds can be utilized as quantum sensors for measuring various physical parameters, particularly magnetic and electric fields, as well as temperature. Due to their small size and possible surface functionalization, fluorescent nanodiamonds are extremely attractive systems for biological and medical applications since they can be used for intracellular experiments. This review focuses on fluorescent nanodiamonds for thermometry with high sensitivity and a nanoscale spatial resolution for the investigation of living systems. The current state of the art, possible further development, and potential limitations of fluorescent nanodiamonds as thermometers will be discussed here.
Collapse
Affiliation(s)
- Anna Ermakova
- Physics Department, Hasselt University, Wetenschapspark 1, 3590 Diepenbeek, Belgium;
- Department of Magnetosphere-Ionosphere Coupling, Royal Belgian Institute for Space Aeronomy, 1180 Brussels, Belgium
| |
Collapse
|
23
|
Javaid S, Qureshi IZ, Khurshid A, Afsar T, Husain FM, Khurshid M, Trembley JH, Razak S. Photoactive metabolite mediated photodynamic therapy of Rhabdomyosarcoma cell lines using medicinal plants and Doxorubicin co-treatments. BMC Complement Med Ther 2024; 24:270. [PMID: 39010043 PMCID: PMC11251096 DOI: 10.1186/s12906-024-04575-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 07/02/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Medicinal plant-mediated combinational therapies have gained importance globally due to minimal side effects and enhanced treatment outcomes compared to single-drug modalities. We aimed to analyze the cytotoxic potential of each conventional treatment i.e., photodynamic therapy (PDT), chemotherapy (doxorubicin hydrochloride; Dox-HCl) with or without various concentrations of medicinal plant extracts (PE) on soft tissue cancer Rhabdomyosarcoma (RD) cell line. METHODS The Rhabdomyosarcoma (RD) cell line was cultured and treated with Photosensitizer (Photosense (AlPc4)), Chemo (Dox-HCl), and their combinations with different concentrations of each plant extract i.e., Thuja occidentalis, Moringa oleifera, Solanum surattense. For the source of illumination, a Diode laser (λ = 630 nm ± 1 nm, Pmax = 1.5 mW) was used. Photosensitizer uptake time (∼ 45 min) was optimized through spectrophotometric measurements (absorption spectroscopy). Drug response of each treatment arm was assessed post 24 h of administration using 3-(4, 5-dimethyl-2-thiazolyl)-2, 5- 5-diphenyl-2 H- tetrazolium bromide (MTT) assay. RESULTS PE-mediated Chemo-Photodynamic therapy (PDT) exhibited synergistic effects (CI < 1). Moreover, Rhabdomyosarcoma culture pretreated with various plant extracts for 24 h exhibited significant inhibition of cell viability however most effective outcomes were shown by low and high doses of Moringa oleifera compared to other plant extracts. Post low doses treated culture with all plant extracts followed by PDT came up with more effectiveness when compared to all di-therapy treatments. CONCLUSION The general outcome of this work shows that the ethanolic plant extracts (higher doses) promote the death of cancerous cells in a dose-dependent way and combining Dox-HCl and photo-mediated photodynamic therapy can yield better therapeutic outcomes.
Collapse
Affiliation(s)
- Sumbal Javaid
- Animal Physiology Laboratory, Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Biophotonics and Photonanomedicine Research laboratory (BPRL), Department of Physics and Applied Mathematics, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
| | - Irfan Zia Qureshi
- Animal Physiology Laboratory, Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Ahmat Khurshid
- Animal Physiology Laboratory, Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Biophotonics and Photonanomedicine Research laboratory (BPRL), Department of Physics and Applied Mathematics, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
| | - Tayyaba Afsar
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, KSA, 11451, Saudi Arabia
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Khurshid
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Janeen H Trembley
- Minneapolis VA Health Care System Research Service, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Suhail Razak
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, KSA, 11451, Saudi Arabia.
| |
Collapse
|
24
|
Abd-El-Aziz A, Ahmed SA, Zhang X, Ma N, Abd-El-Aziz AS. Macromolecules incorporating transition metals in the treatment and detection of cancer and infectious diseases: Progress over the last decade. Coord Chem Rev 2024; 510:215732. [DOI: 10.1016/j.ccr.2024.215732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
25
|
Shi H, Carter OWL, Ponte F, Imberti C, Gomez-Gonzalez MA, Cacho-Nerin F, Quinn PD, Parker JE, Sicilia E, Huang H, Sadler PJ. A Photodynamic and Photochemotherapeutic Platinum-Iridium Charge-Transfer Conjugate for Anticancer Therapy. Angew Chem Int Ed Engl 2024; 63:e202400476. [PMID: 38656762 DOI: 10.1002/anie.202400476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Indexed: 04/26/2024]
Abstract
The novel hetero-dinuclear complex trans,trans,trans-[PtIV(py)2(N3)2(OH)(μ-OOCCH2CH2CONHCH2-bpyMe)IrIII(ppy)2]Cl (Pt-Ir), exhibits charge transfer between the acceptor photochemotherapeutic Pt(IV) (Pt-OH) and donor photodynamic Ir(III) (Ir-NH2) fragments. It is stable in the dark, but undergoes photodecomposition more rapidly than the Pt(IV) parent complex (Pt-OH) to generate Pt(II) species, an azidyl radical and 1O2. The Ir(III)* excited state, formed after irradiation, can oxidise NADH to NAD⋅ radicals and NAD+. Pt-Ir is highly photocytotoxic towards cancer cells with a high photocytotoxicity index upon irradiation with blue light (465 nm, 4.8 mW/cm2), even with short light-exposure times (10-60 min). In contrast, the mononuclear Pt-OH and Ir-NH2 subunits and their simple mixture are much less potent. Cellular Pt accumulation was higher for Pt-Ir compared to Pt-OH. Irradiation of Pt-Ir in cancer cells damages nuclei and releases chromosomes. Synchrotron-XRF revealed ca. 4× higher levels of intracellular platinum compared to iridium in Pt-Ir treated cells under dark conditions. Luminescent Pt-Ir distributes over the whole cell and generates ROS and 1O2 within 1 h of irradiation. Iridium localises strongly in small compartments, suggestive of complex cleavage and excretion via recycling vesicles (e.g. lysosomes). The combination of PDT and PACT motifs in one molecule, provides Pt-Ir with a novel strategy for multimodal phototherapy.
Collapse
Affiliation(s)
- Huayun Shi
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, U.K
| | - Oliver W L Carter
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, U.K
| | - Fortuna Ponte
- Department of Chemistry and Chemical Technologies, University of Calabria, via Pietro Bucci, 87036, Arcavacata Rende, Cs, Italy
| | - Cinzia Imberti
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, U.K
| | | | - Fernando Cacho-Nerin
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, U.K
| | - Paul D Quinn
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, U.K
| | - Julia E Parker
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, U.K
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, University of Calabria, via Pietro Bucci, 87036, Arcavacata Rende, Cs, Italy
| | - Huaiyi Huang
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, U.K
- School of Pharmaceutical Science (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China
| | - Peter J Sadler
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, U.K
| |
Collapse
|
26
|
Rehan F, Zhang M, Fang J, Greish K. Therapeutic Applications of Nanomedicine: Recent Developments and Future Perspectives. Molecules 2024; 29:2073. [PMID: 38731563 PMCID: PMC11085487 DOI: 10.3390/molecules29092073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
The concept of nanomedicine has evolved significantly in recent decades, leveraging the unique phenomenon known as the enhanced permeability and retention (EPR) effect. This has facilitated major advancements in targeted drug delivery, imaging, and individualized therapy through the integration of nanotechnology principles into medicine. Numerous nanomedicines have been developed and applied for disease treatment, with a particular focus on cancer therapy. Recently, nanomedicine has been utilized in various advanced fields, including diagnosis, vaccines, immunotherapy, gene delivery, and tissue engineering. Multifunctional nanomedicines facilitate concurrent medication delivery, therapeutic monitoring, and imaging, allowing for immediate responses and personalized treatment plans. This review concerns the major advancement of nanomaterials and their potential applications in the biological and medical fields. Along with this, we also mention the various clinical translations of nanomedicine and the major challenges that nanomedicine is currently facing to overcome the clinical translation barrier.
Collapse
Affiliation(s)
- Farah Rehan
- Department of Molecular Medicine, Al-Jawhara Centre for Molecular Medicine, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 323, Bahrain;
| | - Mingjie Zhang
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan;
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jun Fang
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan;
| | - Khaled Greish
- Department of Molecular Medicine, Al-Jawhara Centre for Molecular Medicine, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 323, Bahrain;
| |
Collapse
|
27
|
Turkmen Koc SN, Rezaei Benam S, Aral IP, Shahbazi R, Ulubayram K. Gold nanoparticles-mediated photothermal and photodynamic therapies for cancer. Int J Pharm 2024; 655:124057. [PMID: 38552752 DOI: 10.1016/j.ijpharm.2024.124057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
Cancer remains one of the major causes of death globally, with one out of every six deaths attributed to the disease. The impact of cancer is felt on psychological, physical, and financial levels, affecting individuals, communities, and healthcare institutions. Conventional cancer treatments have many challenges and inadequacies. Nanomedicine, however, presents a promising solution by not only overcoming these problems but also offering the advantage of combined therapy for treatment-resistant cancers. Nanoparticles specifically engineered for use in nanomedicine can be efficiently targeted to cancer cells through a combination of active and passive techniques, leading to superior tumor-specific accumulation, enhanced drug availability, and reduced systemic toxicity. Among various nanoparticle formulations designed for cancer treatment, gold nanoparticles have gained prominence in the field of nanomedicine due to their photothermal, photodynamic, and immunologic effects without the need for photosensitizers or immunotherapeutic agents. To date, there is no comprehensive literature review that focuses on the photothermal, photodynamic, and immunologic effects of gold nanoparticles. In this review, significant attention has been devoted to examining the parameters pertaining to the structure of gold nanoparticles and laser characteristics, which play a crucial role in influencing the efficacy of photothermal therapy (PTT) and photodynamic therapy (PDT). Moreover, this article provides insights into the success of PTT and PDT mediated by gold nanoparticles in primary cancer treatment, as well as the immunological effects of PTT and PDT on metastasis and recurrence, providing a promising strategy for cancer therapy. In summary, gold nanoparticles, with their unique properties, have the potential for clinical application in various cancer therapies, including the treatment of primary cancer, recurrence and metastasis.
Collapse
Affiliation(s)
- Seyma Nur Turkmen Koc
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Ankara, Türkiye
| | - Sanam Rezaei Benam
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Ipek Pınar Aral
- Department of Radiation Oncology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara Bilkent City Hospital, Ankara, Türkiye
| | - Reza Shahbazi
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA; Tumor Microenvironment & Metastasis, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, USA; Brown Center for Immunotherapy, Indiana University School of Medicine, Indianapolis, USA.
| | - Kezban Ulubayram
- Department of Nanotechnology and Nanomedicine, Hacettepe University, Ankara, Türkiye; Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, Türkiye; Department of Bioengineering, Hacettepe University, Ankara, Türkiye.
| |
Collapse
|
28
|
Wang C, Gu B, Qi S, Hu S, Wang Y. Boosted photo-immunotherapy via near-infrared light excited phototherapy in tumor sites and photo-activation in sentinel lymph nodes. NANOSCALE ADVANCES 2024; 6:2075-2087. [PMID: 38633053 PMCID: PMC11019502 DOI: 10.1039/d4na00032c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/28/2024] [Indexed: 04/19/2024]
Abstract
Phototherapy is a promising modality that could eradicate tumor and trigger immune responses via immunogenic cell death (ICD) to enhance anti-tumor immunity. However, due to the lack of deep-tissue-excitable phototherapeutic agents and appropriate excitation strategies, the utility of phototherapy for efficient activation of the immune system is challenging. Herein, we report functionalized ICG nanoparticles (NPs) with the capture capability of tumor-associated antigens (TAAs). Under near-infrared (NIR) light excitation, the ICG NPs exhibited high-performance phototherapy, i.e., synergistic photothermal therapy and photodynamic therapy, thereby efficiently eradicating primary solid tumor and inducing ICD and subsequently releasing TAAs. The ICG NPs also captured TAAs and delivered them to sentinel lymph nodes, and then the sentinel lymph nodes were activated with NIR light to trigger efficient T-cell immune responses through activation of dendritic cells with the assistance of ICG NP generated reactive oxygen species, inhibiting residual primary tumor recurrence and controlling distant tumor growth. The strategy of NIR light excited phototherapy in tumor sites and photo-activation in sentinel lymph nodes provides a powerful platform for active immune systems for anti-tumor photo-immunotherapy.
Collapse
Affiliation(s)
- Chen Wang
- Med-X Research Institute & School of Biomedical Engineering, Shanghai Jiao Tong University 1954 Huashan Road Shanghai 200030 China
| | - Bobo Gu
- Med-X Research Institute & School of Biomedical Engineering, Shanghai Jiao Tong University 1954 Huashan Road Shanghai 200030 China
| | - Shuhong Qi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology Wuhan Hubei 430074 China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology Wuhan Hubei 430074 China
| | - Siyi Hu
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences Suzhou 215163 China
| | - Yu Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University Shanghai 200092 China
| |
Collapse
|
29
|
Wang W, Niu Y, Zhang N, Wan Y, Xiao Y, Zhao L, Zhao B, Chen W, Huang D. Cascade-Catalyzed Nanogel for Amplifying Starvation Therapy by Nitric Oxide-Mediated Hypoxia Alleviation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:17313-17322. [PMID: 38534029 DOI: 10.1021/acsami.4c01866] [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: 03/28/2024]
Abstract
Glucose oxidase (Gox)-mediated starvation therapy offers a prospective advantage for malignancy treatment by interrupting the glucose supply to neoplastic cells. However, the negative charge of the Gox surface hinders its enrichment in tumor tissues. Furthermore, Gox-mediated starvation therapy infiltrates large amounts of hydrogen peroxide (H2O2) to surround normal tissues and exacerbate intracellular hypoxia. In this study, a cascade-catalyzed nanogel (A-NE) was developed to boost the antitumor effects of starvation therapy by glucose consumption and cascade reactive release of nitric oxide (NO) to relieve hypoxia. First, the surface cross-linking structure of A-NE can serve as a bioimmobilization for Gox, ensuring Gox stability while improving the encapsulation efficiency. Then, Gox-mediated starvation therapy efficiently inhibited the proliferation of tumor cells while generating large amounts of H2O2. In addition, covalent l-arginine (l-Arg) in A-NE consumed H2O2 derived from glucose decomposition to generate NO, which augmented starvation therapy on metastatic tumors by alleviating tumor hypoxia. Eventually, both in vivo and in vitro studies indicated that nanogels remarkably inhibited in situ tumor growth and hindered metastatic tumor recurrence, offering an alternative possibility for clinical intervention.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yafan Niu
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Ni Zhang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yuqing Wan
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yiqing Xiao
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Lingzhi Zhao
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Bingbing Zhao
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Wei Chen
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Dechun Huang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
30
|
Xie Z, Cao B, Zhao J, Liu M, Lao Y, Luo H, Zhong Z, Xiong X, Wei W, Zou T. Ion Pairing Enables Targeted Prodrug Activation via Red Light Photocatalysis: A Proof-of-Concept Study with Anticancer Gold Complexes. J Am Chem Soc 2024; 146:8547-8556. [PMID: 38498689 DOI: 10.1021/jacs.4c00408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Photocatalysis has found increasing applications in biological systems, for example, in localized prodrug activation; however, high-energy light is usually required without giving sufficient efficiency and target selectivity. In this work, we report that ion pairing between photocatalysts and prodrugs can significantly improve the photoactivation efficiency and enable tumor-targeted activation by red light. This is exemplified by a gold-based prodrug (1d) functionalized with a morpholine moiety. Such a modification causes 1d to hydrolyze in aqueous solution, forming a cationic species that tightly interacts with anionic photosensitizers including Eosin Y (EY) and Rose Bengal (RB), along with a significant bathochromic shift of absorption tailing to the far-red region. As a result, a high photoactivation efficiency of 1d by EY or RB under low-energy light was found, leading to an effective release of active gold species in living cells, as monitored by a gold-specific biosensor (GolS-mCherry). Importantly, the morpholine moiety, with pKa ∼6.9, in 1d brings in a highly pH-sensitive and preferential ionic interaction under a slightly acidic condition over the normal physiological pH, enabling tumor-targeted prodrug activation by red light irradiation in vitro and in vivo. Since a similar absorption change was found in other morpholine/amine-containing clinic drugs, photocages, and precursors of reactive labeling intermediates, it is believed that the ion-pairing strategy could be extended for targeted activation of different prodrugs and for mapping of an acidic microenvironment by low-energy light.
Collapse
Affiliation(s)
- Zhiying Xie
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Bei Cao
- Warshel Institute for Computational Biology, and General Education Division, The Chinese University of Hong Kong, Shenzhen 518172, China
- School of Education Sciences, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511453, China
| | - Jing Zhao
- State Key Laboratory of Coordination Chemistry, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Moyi Liu
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yuhan Lao
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Hejiang Luo
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zhi Zhong
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xiaolin Xiong
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Wei Wei
- State Key Laboratory of Coordination Chemistry, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Taotao Zou
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou 510632, China
| |
Collapse
|
31
|
Mehrotra N, Pal K. Tumor targeted nanohybrid for dual stimuli responsive and NIR amplified photothermal/photo-induced thermodynamic/chemodynamic combination therapy. Biomed Mater 2024; 19:035019. [PMID: 38471148 DOI: 10.1088/1748-605x/ad330f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/12/2024] [Indexed: 03/14/2024]
Abstract
The combination of photodynamic (PDT) and chemodynamic therapy (CDT) for cancer treatment has gathered a lot of attention in recent years. However, its efficacy is severely limited by elevated levels of hypoxia and glutathione (GSH) in the tumor microenvironment (TME). Multifunctional nanoparticles that can help remodel the TME while facilitating PDT/CDT combination therapy are the need of the hour. To this effect, we have developed O2self-supplying, free radical generating nanohybrids that exhibit near infra-red (NIR) triggered photothermal (PTT)/photo-induced thermodynamic (P-TDT) and CDT for efficient breast cancer treatment. The surface of nanohybrids has been further modified by biointerfacing with cancer cell membrane. The biomimetic nanohybrids have been comprehensively characterized and found to exhibit high 2,2'-azobis-[2-(2-imidazolin-2-yl)propane] dihydrochloride (AIPH) loading, GSH depletion, oxygen self-supply with TME responsive AIPH release. Biological activity assays demonstrate efficient cellular uptake with homotypic targeting, excellent hemo- and cytocompatibility as well as high intracellular reactive oxygen species generation with synergistic cytotoxicity against tumor cells. The multifunctional nanohybrid proposed in the present study provides an attractive strategy for achieving NIR responsive, tumor targeted PTT/P-TDT/CDT combination therapy for breast cancer treatment.
Collapse
Affiliation(s)
- Neha Mehrotra
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Kaushik Pal
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India
- Department of Mechanical and Industrial Engineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| |
Collapse
|
32
|
Huang W, Zhang L, Sun J, Sun Y, Gong L, Ge S, Zheng Y, Gao W, Wei X. Hypoxia Reversion by Low-Immunogenic Ultra-Acid-Sensitive Comicelles of Protein-Polymer Conjugates Sensitizes Tumors to Photodynamic Therapy. J Am Chem Soc 2024; 146:7543-7554. [PMID: 38469664 DOI: 10.1021/jacs.3c13501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Hypoxia is characteristic of the tumor microenvironment, which is correlated with resistance to photodynamic therapy (PDT), radiotherapy, chemotherapy, and immunotherapy. Catalase is potentially useful to catalyze the conversion of endogenous H2O2 to O2 for hypoxia reversion. However, the efficient delivery of catalase into the hypoxia regions of tumors is a huge challenge. Here, we report the self-assembly of ultra-acid-sensitive polymer conjugates of catalase and albumin into nanomicelles that are responsive to the acidic tumor microenvironment. The immunogenicity of catalase is mitigated by the presence of albumin, which reduces the cross-linking of catalase with B cell receptors, resulting in improved pharmacokinetics. The ultra acid sensitivity of the nanomicelles makes it possible to efficiently escape the lysosomal degradation after endocytosis and permeate into the interior of tumors to reverse hypoxia in vitro and in vivo. In mice bearing triple-negative breast cancer, the nanomicelles loaded with a photosensitizer effectively accumulate and penetrate into the whole tumors to generate a sufficient amount of O2 to reverse hypoxia, leading to enhanced efficacy of PDT without detectable side effects. These findings provide a general strategy of self-assembly to design low-immunogenic ultra-acid-sensitive comicelles of protein-polymer conjugates to reverse tumor hypoxia, which sensitizes tumors to PDT.
Collapse
Affiliation(s)
- Wenchao Huang
- Institute of Medical Technology and Cancer Hospital, Peking University, Beijing 100191, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
- Biomedical Engineering Department, Peking University, Beijing 100191, China
| | - Longshuai Zhang
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
- Biomedical Engineering Department, Peking University, Beijing 100191, China
- Peking University International Cancer Institute, Beijing 100191, China
- Peking University-Yunnan Baiyao International Medical Research Center, Beijing 100191, China
| | - Jiawei Sun
- Institute of Medical Technology and Cancer Hospital, Peking University, Beijing 100191, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
- Biomedical Engineering Department, Peking University, Beijing 100191, China
- Peking University International Cancer Institute, Beijing 100191, China
- Peking University-Yunnan Baiyao International Medical Research Center, Beijing 100191, China
| | - Yuanzi Sun
- Institute of Medical Technology and Cancer Hospital, Peking University, Beijing 100191, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
- Biomedical Engineering Department, Peking University, Beijing 100191, China
- Peking University International Cancer Institute, Beijing 100191, China
- Peking University-Yunnan Baiyao International Medical Research Center, Beijing 100191, China
| | - Like Gong
- Institute of Medical Technology and Cancer Hospital, Peking University, Beijing 100191, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
- Biomedical Engineering Department, Peking University, Beijing 100191, China
- Peking University International Cancer Institute, Beijing 100191, China
- Peking University-Yunnan Baiyao International Medical Research Center, Beijing 100191, China
| | - Sisi Ge
- Institute of Medical Technology and Cancer Hospital, Peking University, Beijing 100191, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
- Biomedical Engineering Department, Peking University, Beijing 100191, China
| | - Yinghao Zheng
- Institute of Medical Technology and Cancer Hospital, Peking University, Beijing 100191, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
- Biomedical Engineering Department, Peking University, Beijing 100191, China
| | - Weiping Gao
- Institute of Medical Technology and Cancer Hospital, Peking University, Beijing 100191, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
- Biomedical Engineering Department, Peking University, Beijing 100191, China
- Peking University International Cancer Institute, Beijing 100191, China
- Peking University-Yunnan Baiyao International Medical Research Center, Beijing 100191, China
| | - Xunbin Wei
- Institute of Medical Technology and Cancer Hospital, Peking University, Beijing 100191, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
- Biomedical Engineering Department, Peking University, Beijing 100191, China
- Peking University International Cancer Institute, Beijing 100191, China
| |
Collapse
|
33
|
Dash BS, Lu YJ, Chen JP. Enhancing Photothermal/Photodynamic Therapy for Glioblastoma by Tumor Hypoxia Alleviation and Heat Shock Protein Inhibition Using IR820-Conjugated Reduced Graphene Oxide Quantum Dots. ACS APPLIED MATERIALS & INTERFACES 2024; 16:13543-13562. [PMID: 38452225 DOI: 10.1021/acsami.3c19152] [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: 03/09/2024]
Abstract
We use low-molecular-weight branched polyethylenimine (PEI) to produce cytocompatible reduced graphene oxide quantum dots (rGOQD) as a photothermal agent and covalently bind it with the photosensitizer IR-820. The rGOQD/IR820 shows high photothermal conversion efficiency and produces reactive oxygen species (ROS) after irradiation with near-infrared (NIR) light for photothermal/photodynamic therapy (PTT/PDT). To improve suspension stability, rGOQD/IR820 was PEGylated by anchoring with the DSPE hydrophobic tails in DSPE-PEG-Mal, leaving the maleimide (Mal) end group for covalent binding with manganese dioxide/bovine serum albumin (MnO2/BSA) and targeting ligand cell-penetrating peptide (CPP) to synthesize rGOQD/IR820/MnO2/CPP. As MnO2 can react with intracellular hydrogen peroxide to produce oxygen for alleviating the hypoxia condition in the acidic tumor microenvironment, the efficacy of PDT could be enhanced by generating more cytotoxic ROS with NIR light. Furthermore, quercetin (Q) was loaded to rGOQD through π-π interaction, which can be released in the endosomes and act as an inhibitor of heat shock protein 70 (HSP70). This sensitizes tumor cells to thermal stress and increases the efficacy of mild-temperature PTT with NIR irradiation. By simultaneously incorporating the HSP70 inhibitor (Q) and the in situ hypoxia alleviating agent (MnO2), the rGOQD/IR820/MnO2/Q/CPP can overcome the limitation of PTT/PDT and enhance the efficacy of targeted phototherapy in vitro. From in vivo study with an orthotopic brain tumor model, rGOQD/IR820/MnO2/Q/CPP administered through tail vein injection can cross the blood-brain barrier and accumulate in the intracranial tumor, after which NIR laser light irradiation can shrink the tumor and prolong the survival times of animals by simultaneously enhancing the efficacy of PTT/PDT to treat glioblastoma.
Collapse
Affiliation(s)
- Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
34
|
Turrini E, Ulfo L, Costantini PE, Saporetti R, Di Giosia M, Nigro M, Petrosino A, Pappagallo L, Kaltenbrunner A, Cantelli A, Pellicioni V, Catanzaro E, Fimognari C, Calvaresi M, Danielli A. Molecular engineering of a spheroid-penetrating phage nanovector for photodynamic treatment of colon cancer cells. Cell Mol Life Sci 2024; 81:144. [PMID: 38494579 PMCID: PMC10944812 DOI: 10.1007/s00018-024-05174-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/17/2024] [Accepted: 01/30/2024] [Indexed: 03/19/2024]
Abstract
Photodynamic therapy (PDT) represents an emerging strategy to treat various malignancies, including colorectal cancer (CC), the third most common cancer type. This work presents an engineered M13 phage retargeted towards CC cells through pentavalent display of a disulfide-constrained peptide nonamer. The M13CC nanovector was conjugated with the photosensitizer Rose Bengal (RB), and the photodynamic anticancer effects of the resulting M13CC-RB bioconjugate were investigated on CC cells. We show that upon irradiation M13CC-RB is able to impair CC cell viability, and that this effect depends on i) photosensitizer concentration and ii) targeting efficiency towards CC cell lines, proving the specificity of the vector compared to unmodified M13 phage. We also demonstrate that M13CC-RB enhances generation and intracellular accumulation of reactive oxygen species (ROS) triggering CC cell death. To further investigate the anticancer potential of M13CC-RB, we performed PDT experiments on 3D CC spheroids, proving, for the first time, the ability of engineered M13 phage conjugates to deeply penetrate multicellular spheroids. Moreover, significant photodynamic effects, including spheroid disruption and cytotoxicity, were readily triggered at picomolar concentrations of the phage vector. Taken together, our results promote engineered M13 phages as promising nanovector platform for targeted photosensitization, paving the way to novel adjuvant approaches to fight CC malignancies.
Collapse
Affiliation(s)
- Eleonora Turrini
- Dipartimento di Scienze per la Qualità della Vita (QUVI), Alma Mater Studiorum, Università Di Bologna, C.So D'Augusto, 237, 47921, Rimini, Italy
| | - Luca Ulfo
- Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum, Università Di Bologna, Via Francesco Selmi 3, 40126, Bologna, Italy
| | - Paolo Emidio Costantini
- Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum, Università Di Bologna, Via Francesco Selmi 3, 40126, Bologna, Italy
| | - Roberto Saporetti
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum, Università Di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy
| | - Matteo Di Giosia
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum, Università Di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy
| | - Michela Nigro
- Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum, Università Di Bologna, Via Francesco Selmi 3, 40126, Bologna, Italy
| | - Annapaola Petrosino
- Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum, Università Di Bologna, Via Francesco Selmi 3, 40126, Bologna, Italy
| | - Lucia Pappagallo
- Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum, Università Di Bologna, Via Francesco Selmi 3, 40126, Bologna, Italy
| | - Alena Kaltenbrunner
- Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum, Università Di Bologna, Via Francesco Selmi 3, 40126, Bologna, Italy
| | - Andrea Cantelli
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum, Università Di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza" Unit of Bologna, Bologna, Italy
| | - Valentina Pellicioni
- Dipartimento di Scienze per la Qualità della Vita (QUVI), Alma Mater Studiorum, Università Di Bologna, C.So D'Augusto, 237, 47921, Rimini, Italy
| | - Elena Catanzaro
- Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Carmela Fimognari
- Dipartimento di Scienze per la Qualità della Vita (QUVI), Alma Mater Studiorum, Università Di Bologna, C.So D'Augusto, 237, 47921, Rimini, Italy
| | - Matteo Calvaresi
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum, Università Di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy.
- Interdepartmental Center for Industrial Research (CIRI-SDV), Health Sciences and Technologies, University of Bologna, Bologna, Italy.
| | - Alberto Danielli
- Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum, Università Di Bologna, Via Francesco Selmi 3, 40126, Bologna, Italy.
- Interdepartmental Center for Industrial Research (CIRI-SDV), Health Sciences and Technologies, University of Bologna, Bologna, Italy.
| |
Collapse
|
35
|
Khafaga AF, Gaballa MMS, Karam R, Shoulah SA, Shamma RN, Khalifa NE, Farrag NE, Noreldin AE. Synergistic therapeutic strategies and engineered nanoparticles for anti-vascular endothelial growth factor therapy in cancer. Life Sci 2024; 341:122499. [PMID: 38342375 DOI: 10.1016/j.lfs.2024.122499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/13/2024]
Abstract
Angiogenesis is one of the defining characteristics of cancer. Vascular endothelial growth factor (VEGF) is crucial for the development of angiogenesis. A growing interest in cancer therapy is being caused by the widespread use of antiangiogenic drugs in treating several types of human cancer. However, this therapeutic approach can worsen resistance, invasion, and overall survival. As we proceed, refining combination strategies and addressing the constraint of targeted treatments are paramount. Therefore, major challenges in using novel combinations of antiangiogenic agents with cytotoxic treatments are currently focused on illustrating the potential of synergistic therapeutic strategies, alongside advancements in nanomedicine and gene therapy, present opportunities for more precise interference with angiogenesis pathways and tumor environments. Nanoparticles have the potential to regulate several crucial activities and improve several drug limitations such as lack of selectivity, non-targeted cytotoxicity, insufficient drug delivery at tumor sites, and multi-drug resistance based on their unique features. The goal of this updated review is to illustrate the enormous potential of novel synergistic therapeutic strategies and the targeted nanoparticles as an alternate strategy for t treating a variety of tumors employing antiangiogenic therapy.
Collapse
Affiliation(s)
- Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina 22758, Egypt.
| | - Mohamed M S Gaballa
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt.
| | - Reham Karam
- Department of Virology, Faculty of Veterinary Medicine, Mansoura University, 35511, Egypt.
| | - Salma A Shoulah
- Department of Animal Medicine (Infectious Diseases), Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt.
| | - Rehab N Shamma
- Department of Pharmaceutics & Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Norhan E Khalifa
- Department of Physiology, Faculty of Veterinary Medicine, Matrouh University, Matrouh 51511, Egypt.
| | - Nehal E Farrag
- Faculty of Veterinary Medicine, Alexandria University, Edfina 22758, Egypt.
| | - Ahmed E Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt.
| |
Collapse
|
36
|
Shi H, Ponte F, Grewal JS, Clarkson GJ, Imberti C, Hands-Portman I, Dallmann R, Sicilia E, Sadler PJ. Tuning the photoactivated anticancer activity of Pt(iv) compounds via distant ferrocene conjugation. Chem Sci 2024; 15:4121-4134. [PMID: 38487220 PMCID: PMC10935708 DOI: 10.1039/d3sc03092j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 01/30/2024] [Indexed: 03/17/2024] Open
Abstract
Photoactive prodrugs offer potential for spatially-selective antitumour activity with minimal effects on normal tissues. Excited-state chemistry can induce novel effects on biochemical pathways and combat resistance to conventional drugs. Photoactive metal complexes in particular, have a rich and relatively unexplored photochemistry, especially an ability to undergo facile intersystem crossing and populate triplet states. We have conjugated the photoactive octahedral Pt(iv) complex trans, trans, trans-[Pt(N3)2(OH)2(py)2] to ferrocene to introduce novel features into a candidate photochemotherapeutic drug. The X-ray crystal structure of the conjugate Pt-Fe confirmed the axial coordination of a ferrocene carboxylate, with Pt(iv) and Fe(ii) 6.07 Å apart. The conjugation of ferrocene red-shifted the absorption spectrum and ferrocene behaves as a light antenna allowing charge transfer from iron to platinum, promoting the photoactivation of Pt-Fe with light of longer wavelength. Cancer cellular accumulation is enhanced, and generation of reactive species is catalysed after photoirradiation, introducing ferroptosis as a contribution towards the cell-death mechanism. TDDFT calculations were performed to shed light on the behaviour of Pt-Fe when it is irradiated. Intersystem spin-crossing allows the formation of triplet states centred on both metal atoms. The dissociative nature of triplet states confirms that they can be involved in ligand detachment due to irradiation. The Pt(ii) photoproducts mainly retain the trans-{Pt(py)2}2+fragment. Visible light irradiation gives rise to micromolar activity for Pt-Fe towards ovarian, lung, prostate and bladder cancer cells under both normoxia and hypoxia, and some photoproducts appear to retain Pt(iv)-Fe(ii) conjugation.
Collapse
Affiliation(s)
- Huayun Shi
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| | - Fortuna Ponte
- Department of Chemistry and Chemical Technologies, University of Calabria via Pietro Bucci, 87036 Arcavacata di Rende Cs Italy
| | - Jaspreet S Grewal
- Division of Biomedical Sciences, Warwick Medical School CV4 7AL Coventry UK
| | - Guy J Clarkson
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| | - Cinzia Imberti
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| | | | - Robert Dallmann
- Division of Biomedical Sciences, Warwick Medical School CV4 7AL Coventry UK
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, University of Calabria via Pietro Bucci, 87036 Arcavacata di Rende Cs Italy
| | - Peter J Sadler
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| |
Collapse
|
37
|
Xu T, Mi L, Namulinda T, Yan YJ, Meerovich GA, Reshetov IV, Kogan EA, Chen ZL. Quaternary ammonium cations conjugated 5,15-diaryltetranaphtho[2,3]porphyrins as photosensitizers for photodynamic therapy. Eur J Med Chem 2024; 267:116228. [PMID: 38354521 DOI: 10.1016/j.ejmech.2024.116228] [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: 01/14/2024] [Revised: 02/03/2024] [Accepted: 02/08/2024] [Indexed: 02/16/2024]
Abstract
In quest for new photosensitizers (PSs) with remarkable antitumor photodynamic efficacy, a series of fifteen quaternary ammonium (QA) cations conjugated 5,15-diaryltetranaphtho[2,3]porphyrins (Ar2TNPs) was synthesized and evaluated in vitro and in vivo to understand how variations in the length of the alkoxy group and the kind of QA cations on meso-phenyl influence the photodynamic antitumor activity. All final compounds (I1-5, II1-5, and III1-5) exhibited robust absorption at 729 nm with significant bathochromic shift and high molar extinction coefficients (1.16 × 105-1.41 × 105 M-1 cm-1), as well as other absorptions at 445, 475, 651, and 714 nm for tumors and other diseases of diverse sizes and depths. Upon exposure to 474 nm light, they displayed intense fluorescence emission with fluorescence quantum yields ranging from 0.32 to 0.43. The ability to generate reactive oxygen species (ROS) was also quantified, attaining a maximum rate of up to 0.0961 s-1. The IC50 values of all the compounds regarding phototoxicity and dark toxicity were determined using KYSE-150 cells, and the phototoxicity indices were calculated. Among these compounds, III1 demonstrated the highest phototoxic index with minimal dark toxicity, and suppressed successfully the growth of esophageal carcinoma xenograft with favorable tolerance in vivo. Furthermore, the histological results showed III1-mediated PDT had a significant cytotoxic effect on the tumor. These outcomes underscore the potential of III1 as a highly effective antitumor photosensitizer drug in photodynamic therapy (PDT).
Collapse
Affiliation(s)
- Tao Xu
- Department of Pharmaceutical Science & Technology, College of Biology and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Le Mi
- Department of Pharmaceutical Science & Technology, College of Biology and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Tabbisa Namulinda
- Department of Pharmaceutical Science & Technology, College of Biology and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Yi-Jia Yan
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, 200040, China; Shanghai Xianhui Pharmaceutical Co., Ltd., Shanghai, 201620, China.
| | - Gennady A Meerovich
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, 119991, Russia
| | | | - Evgeniy Altarovna Kogan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119992, Russia
| | - Zhi-Long Chen
- Department of Pharmaceutical Science & Technology, College of Biology and Medical Engineering, Donghua University, Shanghai, 201620, China; Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
38
|
Tang X, Yang T, Yu D, Xiong H, Zhang S. Current insights and future perspectives of ultraviolet radiation (UV) exposure: Friends and foes to the skin and beyond the skin. ENVIRONMENT INTERNATIONAL 2024; 185:108535. [PMID: 38428192 DOI: 10.1016/j.envint.2024.108535] [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: 11/08/2023] [Revised: 01/25/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
Ultraviolet (UV) radiation is ubiquitous in the environment, which has been classified as an established human carcinogen. As the largest and outermost organ of the body, direct exposure of skin to sunlight or UV radiation can result in sunburn, inflammation, photo-immunosuppression, photoaging and even skin cancers. To date, there are tactics to protect the skin by preventing UV radiation and reducing the amount of UV radiation to the skin. Nevertheless, deciphering the essential regulatory mechanisms may pave the way for therapeutic interventions against UV-induced skin disorders. Additionally, UV light is considered beneficial for specific skin-related conditions in medical UV therapy. Recent evidence indicates that the biological effects of UV exposure extend beyond the skin and include the treatment of inflammatory diseases, solid tumors and certain abnormal behaviors. This review mainly focuses on the effects of UV on the skin. Moreover, novel findings of the biological effects of UV in other organs and systems are also summarized. Nevertheless, the mechanisms through which UV affects the human organism remain to be fully elucidated to achieve a more comprehensive understanding of its biological effects.
Collapse
Affiliation(s)
- Xiaoyou Tang
- Medical College of Tibet University, Lasa 850000, China; Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Tingyi Yang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Daojiang Yu
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China
| | - Hai Xiong
- Medical College of Tibet University, Lasa 850000, China; West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China.
| | - Shuyu Zhang
- Medical College of Tibet University, Lasa 850000, China; Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China; NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang 621099, China.
| |
Collapse
|
39
|
Alcázar JJ. Thiophene Stability in Photodynamic Therapy: A Mathematical Model Approach. Int J Mol Sci 2024; 25:2528. [PMID: 38473777 DOI: 10.3390/ijms25052528] [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: 01/24/2024] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Thiophene-containing photosensitizers are gaining recognition for their role in photodynamic therapy (PDT). However, the inherent reactivity of the thiophene moiety toward singlet oxygen threatens the stability and efficiency of these photosensitizers. This study presents a novel mathematical model capable of predicting the reactivity of thiophene toward singlet oxygen in PDT, using Conceptual Density Functional Theory (CDFT) and genetic programming. The research combines advanced computational methods, including various DFT techniques and symbolic regression, and is validated with experimental data. The findings underscore the capacity of the model to classify photosensitizers based on their photodynamic efficiency and safety, particularly noting that photosensitizers with a constant rate 1000 times lower than that of unmodified thiophene retain their photodynamic performance without substantial singlet oxygen quenching. Additionally, the research offers insights into the impact of electronic effects on thiophene reactivity. Finally, this study significantly advances thiophene-based photosensitizer design, paving the way for therapeutic agents that achieve a desirable balance between efficiency and safety in PDT.
Collapse
Affiliation(s)
- Jackson J Alcázar
- Centro de Química Médica, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7780272, Chile
| |
Collapse
|
40
|
Ziani Z, Cobo S, Berthet N, Royal G. Optical modulation of cell nucleus penetration and singlet oxygen release of a switchable platinum complex. iScience 2024; 27:108704. [PMID: 38299025 PMCID: PMC10829881 DOI: 10.1016/j.isci.2023.108704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/06/2023] [Accepted: 12/07/2023] [Indexed: 02/02/2024] Open
Abstract
The activation of anticancer molecules with visible light constitutes an elegant strategy to target tumors and to improve the selectivity of treatments. In this context, we report here a visible-light activatable bis-platinum complex (DHP-Pt2) incorporating an organic photo-switchable ligand based on the dimethyldihydropyrene moiety. Illumination of this metal complex with red light (660 nm) under air readily produces the corresponding endoperoxide form (CPDO2-Pt2). These two metal complexes exhibit different DNA binding properties and, more importantly, we show that only the photogenerated CPDO2-Pt2 is able to penetrate into cancer cell nuclei, where it is then capable of releasing cytotoxic singlet oxygen. This study represents the first proof-of-concept showing that dimethyldihydropyrene derivatives can be used to transport and deliver singlet oxygen into cancer cell nuclei upon visible-light activation.
Collapse
Affiliation(s)
- Zakaria Ziani
- University Grenoble Alpes, CNRS, DCM, 38000 Grenoble, France
| | - Saioa Cobo
- University Grenoble Alpes, CNRS, DCM, 38000 Grenoble, France
| | | | - Guy Royal
- University Grenoble Alpes, CNRS, DCM, 38000 Grenoble, France
| |
Collapse
|
41
|
Labra-Vázquez P, Mudrak V, Tassé M, Mallet-Ladeira S, Sournia-Saquet A, Malval JP, Lacroix PG, Malfant I. Acetylacetonate Ruthenium Nitrosyls: A Gateway to Nitric Oxide Release in Water under Near-Infrared Excitation by Two-Photon Absorption. Inorg Chem 2023. [PMID: 37994054 DOI: 10.1021/acs.inorgchem.3c03355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
A fundamental challenge for phototriggered therapies is to obtain robust molecular frameworks that can withstand biological media. Photoactivatable nitric oxide (NO) releasing molecules (photoNORMs) based on ruthenium nitrosyl (RuNO) complexes are among the most studied systems due to several appealing features that make them attractive for therapeutic applications. Nevertheless, the propensity of the NO ligand to be attacked by nucleophiles frequently manifests as significant instability in water for this class of photoNORMs. Our approach to overcome this limitation involved enhancing the Ru-NO π-backbonding to lower the electrophilicity at the NO by replacing the commonly employed 2,2'-bipyridine (bpy) ligand by an anionic, electron-rich, acetylacetonate (acac). A versatile and convenient synthetic route is developed and applied for the preparation of a large library of RuNO photoNORMs with the general formula [RuNO(tpy)(acac)]2+ (tpy = 2,2':6',2″-terpyridine). A combined theoretical and experimental analysis of the Ru-NO bonding in these complexes is presented, supported by extensive single-crystal X-ray diffraction experiments and by topological analyses of the electron charge density by DFT. The enhanced π-back-bonding, systematically evidenced by several techniques, resulted in a remarkable stability in water for these complexes, where significant NO release efficiencies were recorded. We finally demonstrate the possibility of obtaining sophisticated water-stable multipolar NO-delivery platforms that can be activated in the near-IR region by two-photon absorption (TPA), as demonstrated for an octupolar complex with a TPA cross section of 1530 GM at λ = 800 nm and for which NO photorelease was demonstrated under TPA irradiation in aqueous media.
Collapse
Affiliation(s)
- Pablo Labra-Vázquez
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, F-31077 Toulouse, France
| | - Vladyslav Mudrak
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, F-31077 Toulouse, France
| | - Marine Tassé
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, F-31077 Toulouse, France
| | - Sonia Mallet-Ladeira
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, F-31077 Toulouse, France
| | - Alix Sournia-Saquet
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, F-31077 Toulouse, France
| | - Jean-Pierre Malval
- Institut de Science des Matériaux de Mulhouse CNRS-UMR 7361, Université de Haute Alsace, 15 rue Jean Starcky, 68057 Mulhouse, France
| | - Pascal G Lacroix
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, F-31077 Toulouse, France
| | - Isabelle Malfant
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, F-31077 Toulouse, France
| |
Collapse
|
42
|
Zhou W, Chen S, Ouyang Y, Huang B, Zhang H, Zhang W, Tian J. A supramolecular nanoplatform for imaging-guided phototherapies via hypoxia tumour microenvironment remodeling. Chem Sci 2023; 14:11481-11489. [PMID: 37886080 PMCID: PMC10599481 DOI: 10.1039/d3sc03797e] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/29/2023] [Indexed: 10/28/2023] Open
Abstract
Photodynamic therapy (PDT) has emerged as an invasive and promising antitumour treatment, however, the hypoxia in deep tumour tissues and the poor water-solubility of photosensitizers as bottlenecks greatly hinder PDT efficiency. Herein, a tumour microenvironment (TME) activated supramolecular nanoplatform consisting of the pillar[5]arene-based amphiphilic polymer POPD, the phototherapeutic agent Cy7-CN, respiratory medication atovaquone (ATO) and chemotherapeutic drug pyridinyl camptothecin (CPT-Py) was constructed for imaging-guided hypoxia-ameliorated phototherapies. Owing to host-guest interaction, the photochemical and photophysical properties of cyanine were improved exceedingly due to the suppression of π-π stacking. Triggered by the acidic microenvironment in tumour sites, the supramolecular nanoplatform would dissociate and release CPT-Py and ATO which inhibits mitochondria-associated oxidative phosphorylation (OXPHOS) and encourages more oxygen to be used in enhanced PDT. In vitro and in vivo studies verified that the rational combination of ATO-enhanced PDT and PTT overcame the disadvantages of single phototherapy and formed mutual promotion, and simultaneously sensitized chemotherapeutic drugs, which resulted in high tumour inhibition. It is hoped that the supramolecular nanoplatform could shed light on the development of phototherapeutic agents.
Collapse
Affiliation(s)
- Weijie Zhou
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Suwen Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Yingjie Ouyang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Baoxuan Huang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Hongman Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Weian Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Jia Tian
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| |
Collapse
|
43
|
Yang Y, Huangfu L, Li H, Yang D. Research progress of hyperthermia in tumor therapy by influencing metabolic reprogramming of tumor cells. Int J Hyperthermia 2023; 40:2270654. [PMID: 37871910 DOI: 10.1080/02656736.2023.2270654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Cellular metabolic reprogramming is an important feature of malignant tumors. Metabolic reprogramming causes changes in the levels or types of specific metabolites inside and outside the cell, which affects tumorigenesis and progression by influencing gene expression, the cellular state, and the tumor microenvironment. During tumorigenesis, a series of changes in the glucose metabolism, fatty acid metabolism, amino acid metabolism, and cholesterol metabolism of tumor cells occur, which are involved in the process of cellular carcinogenesis and constitute part of the underlying mechanisms of tumor formation. Hyperthermia, as one of the main therapeutic tools for malignant tumors, has obvious effects on tumor cell metabolism. In this paper, we will combine the latest research progress in the field of cellular metabolic reprogramming and focus on the current experimental research and clinical treatment of hyperthermia in cellular metabolic reprogramming to discuss the feasibility of cellular metabolic reprogramming-related mechanisms guiding hyperthermia in malignant tumor treatment, so as to provide more ideas for hyperthermia to treat malignant tumors through the direction of cellular metabolic reprogramming.
Collapse
Affiliation(s)
- Yuchuan Yang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Linkuan Huangfu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Huizhen Li
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Daoke Yang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|
44
|
Petrosino A, Saporetti R, Starinieri F, Sarti E, Ulfo L, Boselli L, Cantelli A, Morini A, Zadran SK, Zuccheri G, Pasquini Z, Di Giosia M, Prodi L, Pompa PP, Costantini PE, Calvaresi M, Danielli A. A modular phage vector platform for targeted photodynamic therapy of Gram-negative bacterial pathogens. iScience 2023; 26:108032. [PMID: 37822492 PMCID: PMC10563061 DOI: 10.1016/j.isci.2023.108032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/04/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023] Open
Abstract
Growing antibiotic resistance has encouraged the revival of phage-inspired antimicrobial approaches. On the other hand, photodynamic therapy (PDT) is considered a very promising research domain for the protection against infectious diseases. Yet, very few efforts have been made to combine the advantages of both approaches in a modular, retargetable platform. Here, we foster the M13 bacteriophage as a multifunctional scaffold, enabling the selective photodynamic killing of bacteria. We took advantage of the well-defined molecular biology of M13 to functionalize its capsid with hundreds of photo-activable Rose Bengal sensitizers and contemporarily target this light-triggerable nanobot to specific bacterial species by phage display of peptide targeting moieties fused to the minor coat protein pIII of the phage. Upon light irradiation of the specimen, the targeted killing of diverse Gram(-) pathogens occurred at subnanomolar concentrations of the phage vector. Our findings contribute to the development of antimicrobials based on targeted and triggerable phage-based nanobiotherapeutics.
Collapse
Affiliation(s)
- Annapaola Petrosino
- Dipartimento di Farmacia e Biotecnologie (FaBiT) – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy
| | - Roberto Saporetti
- Dipartimento di Chimica “Giacomo Ciamician” – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126 Bologna, Italy
| | - Francesco Starinieri
- Dipartimento di Farmacia e Biotecnologie (FaBiT) – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy
| | - Edoardo Sarti
- Dipartimento di Farmacia e Biotecnologie (FaBiT) – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy
| | - Luca Ulfo
- Dipartimento di Farmacia e Biotecnologie (FaBiT) – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy
| | - Luca Boselli
- Nanobiointeractions and Nanodiagnostics Laboratory, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Andrea Cantelli
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza" Unit of Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Andrea Morini
- Dipartimento di Farmacia e Biotecnologie (FaBiT) – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy
| | - Suleman Khan Zadran
- Dipartimento di Farmacia e Biotecnologie (FaBiT) – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy
| | - Giampaolo Zuccheri
- Dipartimento di Farmacia e Biotecnologie (FaBiT) – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy
- CIRI SDV – Centro Interdipartimentale Scienze della Vita - Alma Mater Studiorum - Università di Bologna, Via Tolara di Sopra, 41/E - 40064 Ozzano dell'Emilia (BO), Italy
| | - Zeno Pasquini
- Infectious Diseases Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Matteo Di Giosia
- Dipartimento di Chimica “Giacomo Ciamician” – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126 Bologna, Italy
| | - Luca Prodi
- Dipartimento di Chimica “Giacomo Ciamician” – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126 Bologna, Italy
- CIRI SDV – Centro Interdipartimentale Scienze della Vita - Alma Mater Studiorum - Università di Bologna, Via Tolara di Sopra, 41/E - 40064 Ozzano dell'Emilia (BO), Italy
| | - Pier Paolo Pompa
- Nanobiointeractions and Nanodiagnostics Laboratory, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Paolo Emidio Costantini
- Dipartimento di Farmacia e Biotecnologie (FaBiT) – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy
| | - Matteo Calvaresi
- Dipartimento di Chimica “Giacomo Ciamician” – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126 Bologna, Italy
- CIRI SDV – Centro Interdipartimentale Scienze della Vita - Alma Mater Studiorum - Università di Bologna, Via Tolara di Sopra, 41/E - 40064 Ozzano dell'Emilia (BO), Italy
| | - Alberto Danielli
- Dipartimento di Farmacia e Biotecnologie (FaBiT) – Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy
- CIRI SDV – Centro Interdipartimentale Scienze della Vita - Alma Mater Studiorum - Università di Bologna, Via Tolara di Sopra, 41/E - 40064 Ozzano dell'Emilia (BO), Italy
| |
Collapse
|
45
|
Pediredla A, Scopelliti MG, Narasimhan S, Chamanzar M, Gkioulekas I. Optimized virtual optical waveguides enhance light throughput in scattering media. Nat Commun 2023; 14:5681. [PMID: 37709758 PMCID: PMC10502147 DOI: 10.1038/s41467-023-40864-z] [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: 08/03/2021] [Accepted: 08/10/2023] [Indexed: 09/16/2023] Open
Abstract
Ultrasonically-sculpted gradient-index optical waveguides enable non-invasive light confinement inside scattering media. The confinement level strongly depends on ultrasound parameters (e.g., amplitude, frequency), and medium optical properties (e.g., extinction coefficient). We develop a physically-accurate simulator, and use it to quantify these dependencies for a radially-symmetric virtual optical waveguide. Our analysis provides insights for optimizing virtual optical waveguides for given applications. We leverage these insights to configure virtual optical waveguides that improve light confinement fourfold compared to previous configurations at five mean free paths. We show that virtual optical waveguides enhance light throughput by 50% compared to an ideal external lens, in a medium with bladder-like optical properties at one transport mean free path. We corroborate these simulation findings with real experiments: we demonstrate, for the first time, that virtual optical waveguides recycle scattered light, and enhance light throughput by 15% compared to an external lens at five transport mean free paths.
Collapse
Affiliation(s)
- Adithya Pediredla
- Carnegie Mellon University, Pittsburgh, PA, USA.
- Dartmouth College, United States of America (work done during Pediredla's time at CMU), Hanover, New Hampshire, USA.
| | | | | | | | | |
Collapse
|
46
|
Kim TE, Chang JE. Recent Studies in Photodynamic Therapy for Cancer Treatment: From Basic Research to Clinical Trials. Pharmaceutics 2023; 15:2257. [PMID: 37765226 PMCID: PMC10535460 DOI: 10.3390/pharmaceutics15092257] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Photodynamic therapy (PDT) is an emerging and less invasive treatment modality for various types of cancer. This review provides an overview of recent trends in PDT research, ranging from basic research to ongoing clinical trials, focusing on different cancer types. Lung cancer, head and neck cancer, non-melanoma skin cancer, prostate cancer, and breast cancer are discussed in this context. In lung cancer, porfimer sodium, chlorin e6, and verteporfin have shown promising results in preclinical studies and clinical trials. For head and neck cancer, PDT has demonstrated effectiveness as an adjuvant treatment after surgery. PDT with temoporfin, redaporfin, photochlor, and IR700 shows potential in early stage larynx cancer and recurrent head and neck carcinoma. Non-melanoma skin cancer has been effectively treated with PDT using methyl aminolevulinate and 5-aminolevulinic acid. In prostate cancer and breast cancer, PDT research is focused on developing targeted photosensitizers to improve tumor-specific uptake and treatment response. In conclusion, PDT continues to evolve as a promising cancer treatment strategy, with ongoing research spanning from fundamental investigations to clinical trials, exploring various photosensitizers and treatment combinations. This review sheds light on the recent advancements in PDT for cancer therapy and highlights its potential for personalized and targeted treatments.
Collapse
Affiliation(s)
| | - Ji-Eun Chang
- College of Pharmacy, Dongduk Women’s University, Seoul 02748, Republic of Korea
| |
Collapse
|
47
|
Rees TW, Ho P, Hess J. Recent Advances in Metal Complexes for Antimicrobial Photodynamic Therapy. Chembiochem 2023; 24:e202200796. [PMID: 36917084 PMCID: PMC10947373 DOI: 10.1002/cbic.202200796] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/16/2023]
Abstract
Antimicrobial resistance (AMR) is a growing global problem with more than 1 million deaths due to AMR infection in 2019 alone. New and innovative therapeutics are required to overcome this challenge. Antimicrobial photodynamic therapy (aPDT) is a rapidly growing area of research poised to provide much needed help in the fight against AMR. aPDT works by administering a photosensitizer (PS) that is activated only when irradiated with light, allowing high spatiotemporal control and selectivity. The PS typically generates reactive oxygen species (ROS), which can damage a variety of key biological targets, potentially circumventing existing resistance mechanisms. Metal complexes are well known to display excellent optoelectronic properties, and recent focus has begun to shift towards their application in tackling microbial infections. Herein, we review the last five years of progress in the emerging field of small-molecule metal complex PSs for aPDT.
Collapse
Affiliation(s)
- Thomas W. Rees
- The Francis Crick Institute1 Midland RoadLondonNW1 1ATUK
| | - Po‐Yu Ho
- The Francis Crick Institute1 Midland RoadLondonNW1 1ATUK
- Department of ChemistryKing's College LondonBritannia House, 7 Trinity StreetLondonSE1 1DBUK
| | - Jeannine Hess
- The Francis Crick Institute1 Midland RoadLondonNW1 1ATUK
- Department of ChemistryKing's College LondonBritannia House, 7 Trinity StreetLondonSE1 1DBUK
| |
Collapse
|
48
|
Lin Y, Yang B, Huang Y, Zhang Y, Jiang Y, Ma L, Shen YQ. Mitochondrial DNA-targeted therapy: A novel approach to combat cancer. CELL INSIGHT 2023; 2:100113. [PMID: 37554301 PMCID: PMC10404627 DOI: 10.1016/j.cellin.2023.100113] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 08/10/2023]
Abstract
Mitochondrial DNA (mtDNA) encodes proteins and RNAs that are essential for mitochondrial function and cellular homeostasis, and participates in important processes of cellular bioenergetics and metabolism. Alterations in mtDNA are associated with various diseases, especially cancers, and are considered as biomarkers for some types of tumors. Moreover, mtDNA alterations have been found to affect the proliferation, progression and metastasis of cancer cells, as well as their interactions with the immune system and the tumor microenvironment (TME). The important role of mtDNA in cancer development makes it a significant target for cancer treatment. In recent years, many novel therapeutic methods targeting mtDNA have emerged. In this study, we first discussed how cancerogenesis is triggered by mtDNA mutations, including alterations in gene copy number, aberrant gene expression and epigenetic modifications. Then, we described in detail the mechanisms underlying the interactions between mtDNA and the extramitochondrial environment, which are crucial for understanding the efficacy and safety of mtDNA-targeted therapy. Next, we provided a comprehensive overview of the recent progress in cancer therapy strategies that target mtDNA. We classified them into two categories based on their mechanisms of action: indirect and direct targeting strategies. Indirect targeting strategies aimed to induce mtDNA damage and dysfunction by modulating pathways that are involved in mtDNA stability and integrity, while direct targeting strategies utilized molecules that can selectively bind to or cleave mtDNA to achieve the therapeutic efficacy. This study highlights the importance of mtDNA-targeted therapy in cancer treatment, and will provide insights for future research and development of targeted drugs and therapeutic strategies.
Collapse
Affiliation(s)
- Yumeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Bowen Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yibo Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - You Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yu Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Longyun Ma
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
49
|
Shi H, Xiong CF, Zhang LJ, Cao HC, Wang R, Pan P, Guo HY, Liu T. Light-Triggered Nitric Oxide Nanogenerator with High l-Arginine Loading for Synergistic Photodynamic/Gas/Photothermal Therapy. Adv Healthc Mater 2023; 12:e2300012. [PMID: 36929147 DOI: 10.1002/adhm.202300012] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/19/2023] [Indexed: 03/18/2023]
Abstract
The development of nanomedicines that combine photothermal therapy (PTT) with photodynamic therapy (PDT) is considered promising for cancer treatment, but still faces the challenge of enhancing tumoricidal efficiency. Fortunately, apart from the well-acknowledged effect on direct tumor cell-killing, nitric oxide (NO) is also considered to be effective for the enhancement of both PTT and PDT. However, both the low loading efficiency of NO precursor and the short half-life time and diffusion distance of NO hamper the synergistic therapeutic efficacy of NO. Taking the aforementioned factors into account, a mitochondria-targeted nitric oxide nanogenerator, EArgFe@Ce6, is constructed to achieve high loading of the NO donor l-Arginine (l-Arg) for synergistic photodynamic/gas/photothermal therapy upon single 660 nm light irradiation. The coordination of epigallocatechin gallate (EGCG) and ferric ions (Fe3+ ) provides EArgFe@Ce6 supreme photothermal capability to perform low-temperature PTT (mPTT). EGCG endows EArgFe@Ce6 with mitochondria-targeting capability and meanwhile favors hypoxia alleviation for enhanced PDT. The PDT-produced massive reactive oxygen species (ROS) further catalyzes l-Arg to generate a considerable amount of NO to perform gas therapy and sensitize both mPTT and PDT. In vitro and in vivo studies demonstrate that the synergistic photodynamic/gas/photothermal therapy triggered by single 660 nm light irradiation is highly effective for tumor treatments.
Collapse
Affiliation(s)
- Hui Shi
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Cheng-Feng Xiong
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Lin-Jun Zhang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Hu-Chen Cao
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Ru Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Pei Pan
- School of Pharmacy, Anhui Medical University, Hefei, 230032, P. R. China
| | - Hai-Yan Guo
- School of Public Health, Anhui Medical University, Hefei, 230032, P. R. China
| | - Tao Liu
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| |
Collapse
|
50
|
Li F, Cao Y, Kan X, Li D, Li Y, Huang C, Liu P. AS1411-conjugated doxorubicin-loaded silver nanotriangles for targeted chemo-photothermal therapy of breast cancer. Nanomedicine (Lond) 2023; 18:1077-1094. [PMID: 37650546 DOI: 10.2217/nnm-2023-0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
Background: Combination therapy has attracted tremendous interest for its great potential in treating cancers. Materials & methods: Based on chitosan-coated silver nanotriangles, polyethylene glycol, AS1411 aptamer and doxorubicin, a multifunctional nanocomposite (AS1411-DOX-AgNTs) was constructed and characterized. Then the photothermal properties, ability to target breast cancer cells and anti-breast cancer effect of AS1411-DOX-AgNTs were evaluated. Results: AS1411-DOX-AgNTs were successfully fabricated and showed excellent photothermal conversion efficiency, breast cancer cell and tumor targeting ability. Compared with single treatments, the combination of AS1411-DOX-AgNTs with near-infrared irradiation possessed the strongest anti-breast cancer effect in vitro and in vivo. Conclusion: AS1411-DOX-AgNTs hold great potential in targeted DOX delivery and combined chemo-photothermal therapy for breast cancer.
Collapse
Affiliation(s)
- Fan Li
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yuyu Cao
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Xuechun Kan
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Dongdong Li
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yan Li
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Cheng Huang
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Peidang Liu
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
- Jiangsu Key Laboratory for Biomaterials & Devices, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| |
Collapse
|