51
|
Sun FY, Wei X, Cui WB, Guo JF, Li H, Zou LY, Ren AM. Theoretical Investigation of Novel Nitrogen-Heterocyclic Iridium(III) Polypyridyl Complexes as Photosensitizers for Two-Photon Photodynamic Therapy. J Med Chem 2024; 67:18157-18169. [PMID: 39367842 DOI: 10.1021/acs.jmedchem.4c01292] [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: 10/07/2024]
Abstract
Two-photon photodynamic therapy (TP-PDT) has become a major cancer treatment due to its larger tissue penetration depth, good spatial selectivity, and less damage to normal cells. In this contribution, a series of novel photosensitizer molecules (Ir-2, Ir-2-1∼Ir-2-4) have been designed based on the experimentally demonstrated photosensitizer [Ir(ppy)2(osip)] (PF6) by fine tuning the π-conjugated structure and introducing different nitrogen-heterocyclic substituents. The electronic structures, one- and two-photon absorption spectra, triplet excited state lifetime, solvation-free energy, and photosensitizing performance were evaluated by means of density functional theory (DFT) and time-dependent density functional theory (TDDFT). The results suggested that the molecule Ir-2, incorporating thiophene as the π-connecting group, exhibits a higher probability of triplet state formation, enhanced two-photon absorption cross-section, and prolonged triplet state lifetime. Furthermore, the four designed nitrogen-heterocyclic complexes Ir-2-1∼Ir-2-4 demonstrate favorable photosensitizing properties, with two-photon absorption cross-sections reaching up to 110 GM and triplet excited state lifetimes exceeding 1000 μs for Ir-2-4.
Collapse
Affiliation(s)
- Feng-Yi Sun
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University College of Chemistry, Changchun 130061, PR China
| | - Xue Wei
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University College of Chemistry, Changchun 130061, PR China
| | - Wei-Bo Cui
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University College of Chemistry, Changchun 130061, PR China
| | - Jing-Fu Guo
- School of Physics, Northeast Normal University, Changchun 130024, PR China
| | - Hui Li
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University College of Chemistry, Changchun 130061, PR China
| | - Lu-Yi Zou
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University College of Chemistry, Changchun 130061, PR China
| | - Ai-Min Ren
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University College of Chemistry, Changchun 130061, PR China
| |
Collapse
|
52
|
Meng X, Zhao N, Zhao D, Zhao H, Wang M, Zhao T, Man S, Dai Y, Zhao Y. Mitigating the skin phototoxicity of sonodynamic therapy via singlet oxygen-consuming metal-organic frameworks. J Control Release 2024; 376:303-317. [PMID: 39413848 DOI: 10.1016/j.jconrel.2024.10.020] [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: 07/25/2024] [Revised: 10/02/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Sonodynamic anti-cancer therapy relies on the highly active singlet oxygen to induce potent cell death. However, the non-specific biodistribution of sonosensitizers post systemic administration results in a significant accumulation in the skin, and hence the daylight-induced phototoxicity. Here, we report a smart metal-organic framework-based nanocarrier with titanium dioxide (TiO2) as the sonosensitizer for reduced phototoxicity in the skin. The organic ligand bears the imidazole moiety that can facilely consume singlet oxygen in the skin without compromising the anti-cancer efficacy. The reaction between imidazole moiety and singlet oxygen was confirmed by the density functional theory (DFT). Upon light irradiation, the nanocarrier can significantly reduce the phototoxicity post light irradiation in a range of normal cells in vitro and in a mouse model in vivo. Meanwhile, the ligand contains a disulfide moiety that can deplete glutathione and orchestrate the singlet oxygen-induced toxicity in the CT-26 colon cancer cells. As a result, the nanocarrier showed superior in vivo antitumor efficacy in a CT-26 tumor-bearing mice model, leading to significant suppression of tumor growth and improved animal survival rates. The current work provides a tailored nanoscale particle engineering approach to simultaneously minimize phototoxicity in the skin and sensitize sonodynamic anti-cancer therapy.
Collapse
Affiliation(s)
- Xuan Meng
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Ning Zhao
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Delong Zhao
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Huanyu Zhao
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Mengjiao Wang
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Tianyang Zhao
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Shuli Man
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Yujie Dai
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Yanjun Zhao
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
53
|
Zhou Y, Zou P, Chen X, Chen P, Shi M, Lang J, Chen M. Overcoming Barriers in Photodynamic Therapy Harnessing Nanogenerators Strategies. Int J Biol Sci 2024; 20:5673-5694. [PMID: 39494340 PMCID: PMC11528466 DOI: 10.7150/ijbs.100317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/03/2024] [Indexed: 11/05/2024] Open
Abstract
Photodynamic therapy (PDT) represents a targeted approach for cancer treatment that employs light and photosensitizers (PSs) to induce the generation of reactive oxygen species (ROS). However, PDT faces obstacles including insufficient PS localization, limited light penetration, and treatment resistance. A potential solution lies in nanogenerators (NGs), which function as self-powered systems capable of generating electrical energy. Recent progress in piezoelectric and triboelectric NGs showcases promising applications in cancer research and drug delivery. Integration of NGs with PDT holds the promise of enhancing treatment efficacy by ensuring sustained PS illumination, enabling direct electrical control of cancer cells, and facilitating improved drug administration. This comprehensive review aims to augment our comprehension of PDT principles, explore associated challenges, and underscore the transformative capacity of NGs in conjunction with PDT. By harnessing NG technology alongside PDT, significant advancement in cancer treatment can be realized. Herein, we present the principal findings and conclusions of this study, offering valuable insights into the integration of NGs to overcome barriers in PDT.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Abdominal Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Pingjin Zou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Xingmin Chen
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Ping Chen
- Department of Abdominal Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Min Shi
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Jinyi Lang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Meihua Chen
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| |
Collapse
|
54
|
Bassler MC, Hiller J, Wackenhut F, Zur Oven-Krockhaus S, Frech P, Schmidt F, Kertzscher C, Rammler T, Ritz R, Braun K, Scheele M, Meixner AJ, Brecht M. Fluorescence lifetime imaging unravels the pathway of glioma cell death upon hypericin-induced photodynamic therapy. RSC Chem Biol 2024; 5:d4cb00107a. [PMID: 39421718 PMCID: PMC11474773 DOI: 10.1039/d4cb00107a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Malignant primary brain tumors are a group of highly aggressive and often infiltrating tumors that lack adequate therapeutic treatments to achieve long time survival. Complete tumor removal is one precondition to reach this goal. A promising approach to optimize resection margins and eliminate remaining infiltrative so-called guerilla cells is photodynamic therapy (PDT) using organic photosensitizers that can pass the disrupted blood-brain-barrier and selectively accumulate in tumor tissue. Hypericin fulfills these conditions and additionally offers outstanding photophysical properties, making it an excellent choice as a photosensitizing molecule for PDT. However, the actual hypericin-induced PDT cell death mechanism is still under debate. In this work, hypericin-induced PDT was investigated by employing the three distinct fluorescent probes hypericin, resorufin and propidium iodide (PI) in fluorescence-lifetime imaging microscopy (FLIM). This approach enables visualizing the PDT-induced photodamaging and dying of single, living glioma cells, as an in vitro tumor model for glioblastoma. Hypericin PDT and FLIM image acquisition were simultaneously induced by 405 nm laser irradiation and sequences of FLIM images and fluorescence spectra were recorded to analyze the PDT progression. The reproducibly observed cellular changes provide insight into the mechanism of cell death during PDT and suggest that apoptosis is the initial mechanism followed by necrosis after continued irradiation. These new insights into the mechanism of hypericin PDT of single glioma cells may help to adjust irradiation doses and improve the implementation as a therapy for primary brain tumors.
Collapse
Affiliation(s)
- Miriam C Bassler
- Process Analysis and Technology (PA&T), Reutlingen University Alteburgstr. 150 72762 Reutlingen Germany
- Institute of Physical and Theoretical Chemistry, University of Tübingen Auf der Morgenstelle 18 72076 Tübingen Germany
| | - Jonas Hiller
- Institute of Physical and Theoretical Chemistry, University of Tübingen Auf der Morgenstelle 18 72076 Tübingen Germany
| | - Frank Wackenhut
- Process Analysis and Technology (PA&T), Reutlingen University Alteburgstr. 150 72762 Reutlingen Germany
| | - Sven Zur Oven-Krockhaus
- Institute of Physical and Theoretical Chemistry, University of Tübingen Auf der Morgenstelle 18 72076 Tübingen Germany
| | - Philipp Frech
- Institute of Physical and Theoretical Chemistry, University of Tübingen Auf der Morgenstelle 18 72076 Tübingen Germany
| | - Felix Schmidt
- Institute of Physical and Theoretical Chemistry, University of Tübingen Auf der Morgenstelle 18 72076 Tübingen Germany
| | - Christoph Kertzscher
- Institute of Physical and Theoretical Chemistry, University of Tübingen Auf der Morgenstelle 18 72076 Tübingen Germany
| | - Tim Rammler
- Institute of Physical and Theoretical Chemistry, University of Tübingen Auf der Morgenstelle 18 72076 Tübingen Germany
| | - Rainer Ritz
- Department of Neurosurgery, Schwarzwald-Baar Clinic 78052 Villingen-Schwenningen Germany
| | - Kai Braun
- Institute of Physical and Theoretical Chemistry, University of Tübingen Auf der Morgenstelle 18 72076 Tübingen Germany
| | - Marcus Scheele
- Institute of Physical and Theoretical Chemistry, University of Tübingen Auf der Morgenstelle 18 72076 Tübingen Germany
| | - Alfred J Meixner
- Institute of Physical and Theoretical Chemistry, University of Tübingen Auf der Morgenstelle 18 72076 Tübingen Germany
| | - Marc Brecht
- Process Analysis and Technology (PA&T), Reutlingen University Alteburgstr. 150 72762 Reutlingen Germany
- Institute of Physical and Theoretical Chemistry, University of Tübingen Auf der Morgenstelle 18 72076 Tübingen Germany
| |
Collapse
|
55
|
Merlin JPJ, Crous A, Abrahamse H. Combining Photodynamic Therapy and Targeted Drug Delivery Systems: Enhancing Mitochondrial Toxicity for Improved Cancer Outcomes. Int J Mol Sci 2024; 25:10796. [PMID: 39409125 PMCID: PMC11477455 DOI: 10.3390/ijms251910796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Cancer treatment continues to be a substantial problem due to tumor complexities and persistence, demanding novel therapeutic techniques. This review investigates the synergistic potential of combining photodynamic therapy (PDT) and tailored medication delivery technologies to increase mitochondrial toxicity and improve cancer outcomes. PDT induces selective cellular damage and death by activating photosensitizers (PS) with certain wavelengths of light. However, PDT's efficacy can be hampered by issues such as poor light penetration and a lack of selectivity. To overcome these challenges, targeted drug delivery systems have emerged as a promising technique for precisely delivering therapeutic medicines to tumor cells while avoiding off-target effects. We investigate how these technologies can improve mitochondrial targeting and damage, which is critical for causing cancer cell death. The combination method seeks to capitalize on the advantages of both modalities: selective PDT activation and specific targeted drug delivery. We review current preclinical and clinical evidence supporting the efficacy of this combination therapy, focusing on case studies and experimental models. This review also addresses issues such as safety, distribution efficiency, resistance mechanisms, and costs. The prospects of further research include advances in photodynamic agents and medication delivery technology, with a focus on personalized treatment. In conclusion, combining PDT with targeted drug delivery systems provides a promising frontier in cancer therapy, with the ability to overcome current treatment limits and open the way for more effective, personalized cancer treatments.
Collapse
Affiliation(s)
- J. P. Jose Merlin
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg 2028, South Africa; (A.C.); (H.A.)
| | | | | |
Collapse
|
56
|
Sanz-Villafruela J, Bermejo-Casadesus C, Zafon E, Martínez-Alonso M, Durá G, Heras A, Soriano-Díaz I, Giussani A, Ortí E, Tebar F, Espino G, Massaguer A. Insights into the anticancer photodynamic activity of Ir(III) and Ru(II) polypyridyl complexes bearing β-carboline ligands. Eur J Med Chem 2024; 276:116618. [PMID: 38972079 DOI: 10.1016/j.ejmech.2024.116618] [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: 01/24/2024] [Revised: 05/31/2024] [Accepted: 06/22/2024] [Indexed: 07/09/2024]
Abstract
Ir(III) and Ru(II) polypyridyl complexes are promising photosensitizers (PSs) for photodynamic therapy (PDT) due to their outstanding photophysical properties. Herein, one series of cyclometallated Ir(III) complexes and two series of Ru(II) polypyridyl derivatives bearing three different thiazolyl-β-carboline N^N' ligands have been synthesized, aiming to evaluate the impact of the different metal fragments ([Ir(C^N)2]+ or [Ru(N^N)2]2+) and N^N' ligands on the photophysical and biological properties. All the compounds exhibit remarkable photostability under blue-light irradiation and are emissive (605 < λem < 720 nm), with the Ru(II) derivatives displaying higher photoluminescence quantum yields and longer excited state lifetimes. The Ir PSs display pKa values between 5.9 and 7.9, whereas their Ru counterparts are less acidic (pKa > 9.3). The presence of the deprotonated form in the Ir-PSs favours the generation of reactive oxygen species (ROS) since, according to theoretical calculations, it features a low-lying ligand-centered triplet excited state (T1 = 3LC) with a long lifetime. All compounds have demonstrated anticancer activity. Ir(III) complexes 1-3 exhibit the highest cytotoxicity in dark conditions, comparable to cisplatin. Their activity is notably enhanced by blue-light irradiation, resulting in nanomolar IC50 values and phototoxicity indexes (PIs) between 70 and 201 in different cancer cell lines. The Ir(III) PSs are also activated by green (with PI between 16 and 19.2) and red light in the case of complex 3 (PI = 8.5). Their antitumor efficacy is confirmed by clonogenic assays and using spheroid models. The Ir(III) complexes rapidly enter cells, accumulating in mitochondria and lysosomes. Upon photoactivation, they generate ROS, leading to mitochondrial dysfunction and lysosomal damage and ultimately cell apoptosis. Additionally, they inhibit cancer cell migration, a crucial step in metastasis. In contrast, Ru(II) complex 6 exhibits moderate mitochondrial activity. Overall, Ir(III) complexes 1-3 show potential for selective light-controlled cancer treatment, providing an alternative mechanism to chemotherapy and the ability to inhibit lethal cancer cell dissemination.
Collapse
Affiliation(s)
- Juan Sanz-Villafruela
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain
| | - Cristina Bermejo-Casadesus
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003, Girona, Spain
| | - Elisenda Zafon
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003, Girona, Spain
| | - Marta Martínez-Alonso
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain
| | - Gema Durá
- Universidad de Castilla-La Mancha, Departamento de Química Inorgánica, Orgánica y Bioquímica. Facultad de Químicas, Avda. Camilo J. Cela 10, 13071, Ciudad Real, Spain
| | - Aranzazu Heras
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain
| | - Iván Soriano-Díaz
- Instituto de Ciencia Molecular, Universidad de Valencia, Catedrático José Beltrán 2, 46980, Paterna, Spain
| | - Angelo Giussani
- Instituto de Ciencia Molecular, Universidad de Valencia, Catedrático José Beltrán 2, 46980, Paterna, Spain
| | - Enrique Ortí
- Instituto de Ciencia Molecular, Universidad de Valencia, Catedrático José Beltrán 2, 46980, Paterna, Spain.
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain.
| | - Gustavo Espino
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain.
| | - Anna Massaguer
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003, Girona, Spain.
| |
Collapse
|
57
|
Duterte MMD, Morales NP, Pitiphat W, Puthongking P, Damrongrungruang T. Effects of photodynamic therapy using bisdemethoxycurcumin combined with melatonin or acetyl-melatonin on C. Albicans. Sci Rep 2024; 14:23082. [PMID: 39367128 PMCID: PMC11452606 DOI: 10.1038/s41598-024-74315-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/25/2024] [Indexed: 10/06/2024] Open
Abstract
The current study aims to explore the efficacy of antifungal photodynamic therapy (PDT) on C. albicans biofilms by combining photosensitizers, bisdemethoxycurcumin (BDMC), and melatonin (MLT) or acetyl-melatonin (AcO-MLT). Additionally, the relationship between different types of reactive oxygen species and PDT's antifungal efficacy was investigated. BDMC, MLT and AcO-MLT were applied, alone and in combination, to 48-hour C. albicans biofilm cultures (n = 6/group). Blue and red LED light (250 mW/cm2 with 37.5 J/cm2 for single or 75 J/cm2 for dual photosensitizer groups) were used to irradiate BDMC groups and MLT/AcO-MLT groups, respectively. For combination groups, blue LEDs and subsequently red LEDs were used. Drop plate assays were performed at 0, 1 and 6 h post-treatment. Colony forming units (CFUs) were then counted after 48 h. Hydroxyl radicals and singlet oxygen were measured using fluorescence spectroscopy and electron spin resonance (ESR) spectroscopy. Additionally, cell cytotoxicity was tested on human oral keratinocytes. Significant CFU reductions were observed with combinations 20 µM BDMC + 20 µM AcO-MLT and 60 µM BDMC + 20 µM MLT at 0 and 1 h post-treatment, respectively. Singlet oxygen production increased with the addition of MLT/AcO-MLT and had moderate-substantial correlations with inhibition at all times. Hydroxyl radical production was not significantly different from the control. Additionally, BDMC exhibited subtle cytotoxicity on human oral keratinocytes. PDT using BDMC + MLT or AcO-MLT, with blue and red LED light, effectively inhibits C. albicans biofilm through singlet oxygen generation. Melatonin acts as a photosensitizer in PDT to inhibit fungal infection.
Collapse
Affiliation(s)
- Maria Margarita D Duterte
- Division of Periodontology, Department of Oral Biomedical Sciences, Faculty of Dentistry, Khon Kaen University, 123 Mittraphap Rd, Amphur Muang, Khon Kaen, 40002, Thailand
| | | | - Waranuch Pitiphat
- Division of Dental Public Health, Department of Preventive Dentistry, Faculty of Dentistry, Khon Kaen University, 123 Mittraphap Rd, Amphur Muang, Khon Kaen, 40002, Thailand
| | - Ploenthip Puthongking
- Melatonin Research Group, Khon Kaen University, Khon Kaen, Thailand
- Faculty of Pharmaceutical Sciences, Khon Kaen University, 123 Mittraphap Road, Amphur Muang, Khon Kaen, 40002, Thailand
| | - Teerasak Damrongrungruang
- Melatonin Research Group, Khon Kaen University, Khon Kaen, Thailand.
- Division of Oral Diagnosis, Department of Oral Biomedical Sciences, Faculty of Dentistry, Khon Kaen University, 123 Mittraphap Rd, Amphur Muang, Khon Kaen, 40002, Thailand.
| |
Collapse
|
58
|
Hosik J, Hosikova B, Binder S, Lenobel R, Kolarikova M, Malina L, Dilenko H, Langova K, Bajgar R, Kolarova H. Effects of Zinc Phthalocyanine Photodynamic Therapy on Vital Structures and Processes in Hela Cells. Int J Mol Sci 2024; 25:10650. [PMID: 39408981 PMCID: PMC11476877 DOI: 10.3390/ijms251910650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
This work presents results on the efficiency of newly designed zinc phthalocyanine-mediated photodynamic therapy of both tumoral and nontumoral cell models using the MTT assay. Further detailed examinations of mechanistic and cell biological effects were focused on the HELA cervical cancer cell model. Here, ROS production, changes in the mitochondrial membrane potential, the determination of genotoxicity, and protein changes determined by capillary chromatography and tandem mass spectrometry with ESI were analyzed. The results showed that, in vitro, 5 Jcm-2 ZnPc PDT caused a significant increase in reactive oxygen species. Still, except for superoxide dismutase, the levels of proteins involved in cell response to oxidative stress did not increase significantly. Furthermore, this therapy damaged mitochondrial membranes, which was proven by a more than 70% voltage-dependent channel protein 1 level decrease and by a 65% mitochondrial membrane potential change 24 h post-therapy. DNA impairment was assessed by an increased level of DNA fragmentation, which might be related to the decreased level of DDB1 (decrease in levels of more than 20% 24 h post-therapy), a protein responsible for maintaining genomic integrity and triggering the DNA repair pathways. Considering these results and the low effective concentration (LC50 = 30 nM), the therapy used is a potentially very promising antitumoral treatment.
Collapse
Affiliation(s)
- Jakub Hosik
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.H.); (S.B.); (M.K.); (L.M.); (H.D.); (K.L.); (R.B.); (H.K.)
| | - Barbora Hosikova
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.H.); (S.B.); (M.K.); (L.M.); (H.D.); (K.L.); (R.B.); (H.K.)
| | - Svatopluk Binder
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.H.); (S.B.); (M.K.); (L.M.); (H.D.); (K.L.); (R.B.); (H.K.)
| | - Rene Lenobel
- Laboratory of Growth Regulators, Faculty of Science, Palacky University and Institute of Experimental Botany of the Czech Academy of Sciences, 77900 Olomouc, Czech Republic;
| | - Marketa Kolarikova
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.H.); (S.B.); (M.K.); (L.M.); (H.D.); (K.L.); (R.B.); (H.K.)
| | - Lukas Malina
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.H.); (S.B.); (M.K.); (L.M.); (H.D.); (K.L.); (R.B.); (H.K.)
| | - Hanna Dilenko
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.H.); (S.B.); (M.K.); (L.M.); (H.D.); (K.L.); (R.B.); (H.K.)
| | - Katerina Langova
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.H.); (S.B.); (M.K.); (L.M.); (H.D.); (K.L.); (R.B.); (H.K.)
| | - Robert Bajgar
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.H.); (S.B.); (M.K.); (L.M.); (H.D.); (K.L.); (R.B.); (H.K.)
| | - Hana Kolarova
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (J.H.); (S.B.); (M.K.); (L.M.); (H.D.); (K.L.); (R.B.); (H.K.)
| |
Collapse
|
59
|
Marinho MAG, da Silva Marques M, de Oliveira Vian C, de Moraes Vaz Batista Filgueira D, Horn AP. Photodynamic therapy with curcumin and near-infrared radiation as an antitumor strategy to glioblastoma cells. Toxicol In Vitro 2024; 100:105917. [PMID: 39142446 DOI: 10.1016/j.tiv.2024.105917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/18/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Glioblastoma is a malignant neoplasm that develops in the central nervous system and is characterized by high rates of cell proliferation and invasion, presenting resistance to treatments and a poor prognosis. Photodynamic therapy (PDT) is a therapeutic modality that can be applied in oncological cases and stands out for being less invasive. Photosensitizers (PS) of natural origin gained prominence in PDT. Curcumin (CUR) is a natural compound that has been used in PDT, considered a promising PS. In this work, we evaluated the effects of PDT-mediated CUR and near-infrared radiation (NIR) in glioblastoma cells. Through trypan blue exclusion analysis, we chose the concentration of 5 μM of CUR and the dose of 2 J/cm2 of NIR that showed better responses in reducing the viable cell number in the C6 cell line and did not show cytotoxic/cytostatic effects in the HaCat cell line. Our results show that there is a positive interaction between CUR and NIR as a PDT model since there was an increase in ROS levels, a decrease in cell proliferation, increase in cytotoxicity with cell death by autophagy and necrosis, in addition to the presence of oxidative damage to proteins. These results suggest that the use of CUR and NIR is a promising strategy for the antitumor application of PDT.
Collapse
Affiliation(s)
- Marcelo Augusto Germani Marinho
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil; Laboratório de Cultura Celular, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil.
| | - Magno da Silva Marques
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil; Laboratório de Neurociências, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil
| | - Camila de Oliveira Vian
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil; Laboratório de Neurociências, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil
| | - Daza de Moraes Vaz Batista Filgueira
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil; Laboratório de Cultura Celular, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil
| | - Ana Paula Horn
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil; Laboratório de Neurociências, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil
| |
Collapse
|
60
|
Anvekar MP, Virupaxi SG, Yavagal C, Kulkarni S, Pai R, Patil VVC. Evaluating the efficacy of cavity disinfection using methylene blue dye with 660-nm diode laser on primary molars: An in vivo study. J Indian Soc Pedod Prev Dent 2024; 42:280-285. [PMID: 39798104 DOI: 10.4103/jisppd.jisppd_258_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 10/03/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND AND OBJECTIVES The purpose of this study was to evaluate the antimicrobial activity of methylene blue dye with 660-nm diode laser in cavity disinfection and to compare the total bacterial count in dentinal samples preexcavation, postexcavation, and postdisinfection. The study design was experimental in vivo. MATERIALS AND METHODS Fifteen children aged 5-12 years with 15 primary molars requiring atraumatic restorative treatment (ART) were selected. Dentinal samples were collected from the carious tooth with a sterile spoon excavator three times, i.e., before caries excavation (preexcavation), after hand excavation of caries (postexcavation), and after doing laser photodynamic therapy with methylene blue dye and 660-nm diode laser (postdisinfection) and sent for microbiological analysis. The samples were transported in reduced transport fluid. The total bacterial count was carried out by an experienced microbiologist immediately after reporting. RESULTS A significant number of bacteria was present after hand excavation of caries using ART. The bacterial count significantly reduced postlaser disinfection. The total viable bacterial count was reduced to 87.17% with hand excavation of caries, whereas it was reduced to 98.59% postlaser disinfection using methylene blue dye with 660-nm diode laser. CONCLUSION It was seen that ART cannot eliminate all the microorganisms. Methylene blue dye with a 660-nm diode laser was able to reduce the total bacterial count and can be effectively used as an antimicrobial cavity disinfectant.
Collapse
Affiliation(s)
- Mitaj Pandharinath Anvekar
- Department of Pediatric and Preventive Dentistry, Maratha Mandal's Nathajirao G. Halgekar Institute of Dental Sciences and Research Centre, Belagavi, Karnataka, India
| | | | | | | | | | | |
Collapse
|
61
|
Porubský M, Hodoň J, Stanková J, Džubák P, Hajdúch M, Urban M, Hlaváč J. Near-infrared pH-switchable BODIPY photosensitizers for dual biotin/cRGD targeted photodynamic therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 259:113010. [PMID: 39141981 DOI: 10.1016/j.jphotobiol.2024.113010] [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: 05/28/2024] [Revised: 07/20/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Photodynamic therapy (PDT) is a clinically-approved cancer treatment that is based on production of cytotoxic reactive oxygen species to induce cell death. However, its efficiency depends on distribution of photosensitizer (PS) and depth of light penetration through the tissues. Tendency of pathological cancer tissues to exhibit lower pH than healthy tissues inspired us to explore dual-targeted pH-activatable photosensitizers based on tunable near-infrared (NIR) boron-dipyrromethene (BODIPY) dyes. Our BODIPY PSs were designed to carry three main attributes: (i) biotin or cRGD peptide as an effective cancer cell targeting unit, (ii) amino moiety that is protonated in acidic (pH <6.5) conditions for pH-activation of the PS based on photoinduced electron transfer (PET) and (iii) hydrophilic groups enhancing the water solubility of very hydrophobic BODIPY dyes. Illumination of such compounds with suitable light (>640nm) allowed for high phototoxicity against HeLa (αvβ3 integrin and biotin receptor positive) and A549 (biotin receptor positive) cells compared to healthy MRC-5 (biotin negative) cells. Moreover, no dark toxicity was observed on selected cell lines (>10 μM) providing promising photosensitizers for tumour-targeted photodynamic therapy.
Collapse
Affiliation(s)
- Martin Porubský
- Department of Organic Chemistry, Faculty of Science, Palacký University, Tr. 17. Listopadu 12, 771 46 Olomouc, Czech Republic.
| | - Jiří Hodoň
- Department of Organic Chemistry, Faculty of Science, Palacký University, Tr. 17. Listopadu 12, 771 46 Olomouc, Czech Republic; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 779 00 Olomouc, Czech Republic
| | - Jarmila Stanková
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 779 00 Olomouc, Czech Republic
| | - Petr Džubák
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 779 00 Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 779 00 Olomouc, Czech Republic
| | - Milan Urban
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 779 00 Olomouc, Czech Republic
| | - Jan Hlaváč
- Department of Organic Chemistry, Faculty of Science, Palacký University, Tr. 17. Listopadu 12, 771 46 Olomouc, Czech Republic.
| |
Collapse
|
62
|
Ain QT. Recent development of nanomaterials-based PDT to improve immunogenic cell death. Photochem Photobiol Sci 2024; 23:1983-1998. [PMID: 39320675 DOI: 10.1007/s43630-024-00638-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024]
Abstract
Photodynamic therapy (PDT) is a clinically approved therapeutic modality for treating oncological and non-oncological disorders. PDT has proclaimed multiple benefits over further traditional cancer therapies including its minimal systemic toxicity and selective ability to eliminate irradiated tumors. In PDT, a photosensitizing substance localizes in tumor tissues and becomes active when exposed to a particular wavelength of laser light. This produces reactive oxygen species (ROS), which induce neoplastic cells to die and lead to the regression of tumors. The contributions of ROS to PDT-induced tumor destruction are described by three basic processes including direct or indirect cell death, vascular destruction, and immunogenic cell death. However, the efficiency of PDT is significantly limited by the inherent nature and tumor microenvironment. Combining immunotherapy with PDT has recently been shown to improve tumor immunogenicity while decreasing immunoregulatory repression, thereby gently modifying the anticancer immune response with long-term immunological memory effects. This review highlights the fundamental ideas, essential elements, and mechanisms of PDT as well as nanomaterial-based PDT to boost tumor immunogenicity. Moreover, the synergistic use of immunotherapy in combination with PDT to enhance immune responses against tumors is emphasized.
Collapse
Affiliation(s)
- Qura Tul Ain
- Department of Physics, The Women University Multan, Khawajabad, Multan, Pakistan.
| |
Collapse
|
63
|
Hou W, Shen L, Zhu Y, Wang X, Du T, Yang F, Zhu Y. Fullerene Derivatives for Tumor Treatment: Mechanisms and Application. Int J Nanomedicine 2024; 19:9771-9797. [PMID: 39345909 PMCID: PMC11430870 DOI: 10.2147/ijn.s476601] [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: 05/02/2024] [Accepted: 08/28/2024] [Indexed: 10/01/2024] Open
Abstract
Fullerenes hold tremendous potential as alternatives to conventional chemotherapy or radiotherapy for tumor treatment due to their abilities to photodynamically kill tumor cells, destroy the tumor vasculature, inhibit tumor metastasis and activate anti-tumor immune responses, while protecting normal tissue through antioxidative effects. The symmetrical hollow molecular structures of fullerenes with abundant C=C bonds allow versatile chemical modification with diverse functional groups, metal clusters and biomacromolecules to synthesize a wide range of fullerene derivatives with increased water solubility, improved biocompatibility, enhanced photodynamic properties and stronger targeting abilities. This review introduces the anti-tumor mechanisms of fullerenes and summarizes the most recent works on the functionalization of fullerenes and the application of fullerene derivatives in tumor treatment. This review aims to serve as a valuable reference for further development and clinical application of anti-tumor fullerene derivatives.
Collapse
Affiliation(s)
- Wenjia Hou
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, People's Republic of China
| | - Lan Shen
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Yimin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Xuanjia Wang
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Tianyu Du
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Fang Yang
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| |
Collapse
|
64
|
Manescu (Paltanea) V, Antoniac I, Paltanea G, Nemoianu IV, Mohan AG, Antoniac A, Rau JV, Laptoiu SA, Mihai P, Gavrila H, Al-Moushaly AR, Bodog AD. Magnetic Hyperthermia in Glioblastoma Multiforme Treatment. Int J Mol Sci 2024; 25:10065. [PMID: 39337552 PMCID: PMC11432100 DOI: 10.3390/ijms251810065] [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: 08/15/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Glioblastoma multiforme (GBM) represents one of the most critical oncological diseases in neurological practice, being considered highly aggressive with a dismal prognosis. At a worldwide level, new therapeutic methods are continuously being researched. Magnetic hyperthermia (MHT) has been investigated for more than 30 years as a solution used as a single therapy or combined with others for glioma tumor assessment in preclinical and clinical studies. It is based on magnetic nanoparticles (MNPs) that are injected into the tumor, and, under the effect of an external alternating magnetic field, they produce heat with temperatures higher than 42 °C, which determines cancer cell death. It is well known that iron oxide nanoparticles have received FDA approval for anemia treatment and to be used as contrast substances in the medical imagining domain. Today, energetic, efficient MNPs are developed that are especially dedicated to MHT treatments. In this review, the subject's importance will be emphasized by specifying the number of patients with cancer worldwide, presenting the main features of GBM, and detailing the physical theory accompanying the MHT treatment. Then, synthesis routes for thermally efficient MNP manufacturing, strategies adopted in practice for increasing MHT heat performance, and significant in vitro and in vivo studies are presented. This review paper also includes combined cancer therapies, the main reasons for using these approaches with MHT, and important clinical studies on human subjects found in the literature. This review ends by describing the most critical challenges associated with MHT and future perspectives. It is concluded that MHT can be successfully and regularly applied as a treatment for GBM if specific improvements are made.
Collapse
Affiliation(s)
- Veronica Manescu (Paltanea)
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (I.A.); (A.A.)
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (I.V.N.)
| | - Iulian Antoniac
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (I.A.); (A.A.)
- Academy of Romanian Scientists, 54 Splaiul Independentei, RO-050094 Bucharest, Romania
| | - Gheorghe Paltanea
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (I.V.N.)
| | - Iosif Vasile Nemoianu
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (I.V.N.)
| | - Aurel George Mohan
- Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania
- Department of Neurosurgery, Clinical Emergency Hospital Oradea, 65 Gheorghe Doja Street, RO-410169 Oradea, Romania
| | - Aurora Antoniac
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (I.A.); (A.A.)
| | - Julietta V. Rau
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (ISM-CNR), Via del Fosso del Cavaliere 100, 00133 Rome, Italy;
- Institute of Pharmacy, Department of Analytical, Physical and Colloid Chemistry, I.M. Sechenov First Moscow State Medical University, Trubetskaya St. 8, Build.2, 119048 Moscow, Russia
| | - Stefan Alexandru Laptoiu
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (I.A.); (A.A.)
| | - Petruta Mihai
- Faculty of Entrepreneurship, Business Engineering and Management, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania;
| | - Horia Gavrila
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (I.V.N.)
- Technical Sciences Academy of Romania, 26 Bulevardul Dacia, RO-030167 Bucharest, Romania
| | | | - Alin Danut Bodog
- Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania
| |
Collapse
|
65
|
V. Cabral F, Xu Q, Greer A, Lyons AM, Hasan T. Superhydrophobic Dressing for Singlet Oxygen Delivery in Antimicrobial Photodynamic Therapy against Multidrug-Resistant Bacterial Biofilms. ACS APPLIED BIO MATERIALS 2024; 7:6175-6185. [PMID: 39166743 PMCID: PMC11409211 DOI: 10.1021/acsabm.4c00733] [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: 06/04/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024]
Abstract
The rise of antimicrobial resistance poses a critical public health threat worldwide. While antimicrobial photodynamic therapy (aPDT) has demonstrated efficacy against multidrug-resistant (MDR) bacteria, its effectiveness can be limited by several factors, including the delivery of the photosensitizer (PS) to the site of interest and the development of bacterial resistance to PS uptake. There is a need for alternative methods, one of which is superhydrophobic antimicrobial photodynamic therapy (SH-aPDT), which we report here. SH-aPDT is a technique that isolates the PS on a superhydrophobic (SH) membrane, generating airborne singlet oxygen (1O2) that can diffuse up to 1 mm away from the membrane. In this study, we developed a SH polydimethylsiloxane dressing coated with PS verteporfin. These dressings contain air channels called a plastron for supplying oxygen for aPDT and are designed so that there is no direct contact of the PS with the tissue. Our investigation focuses on the efficacy of SH-aPDT on biofilms formed by drug-sensitive and MDR strains of Gram-positive (Staphylococcus aureus and S. aureus methicillin-resistant) and Gram-negative bacteria (Pseudomonas aeruginosa and P. aeruginosa carbapenem-resistant). SH-aPDT reduces bacterial biofilms by approximately 3 log with a concomitant decrease in their metabolism as measured by MTT. Additionally, the treatment disrupted extracellular polymeric substances, leading to a decrease in biomass and biofilm thickness. This innovative SH-aPDT approach holds great potential for combating antimicrobial resistance, offering an effective strategy to address the challenges posed by drug-resistant wound infections.
Collapse
Affiliation(s)
- Fernanda V. Cabral
- Wellman
Center for Photomedicine, Massachusetts
General Hospital and Harvard Medical School, 40 Blossom Street, Boston, Massachusetts 02114, United States
| | - QianFeng Xu
- SingletO2
Therapeutics LLC, VentureLink,
Room 524B, 211 Warren Street, Newark, New Jersey 07103, United States
| | - Alexander Greer
- SingletO2
Therapeutics LLC, VentureLink,
Room 524B, 211 Warren Street, Newark, New Jersey 07103, United States
- Ph.D.
Program in Chemistry, The Graduate Center
of the City University of New York, 365 Fifth Avenue, New York, New York 10016, United States
- Department
of Chemistry, Brooklyn College, City University
of New York, Brooklyn, New York 11210, United States
| | - Alan M. Lyons
- SingletO2
Therapeutics LLC, VentureLink,
Room 524B, 211 Warren Street, Newark, New Jersey 07103, United States
- Ph.D.
Program in Chemistry, The Graduate Center
of the City University of New York, 365 Fifth Avenue, New York, New York 10016, United States
- Department
of Chemistry, College of Staten Island, City University of New York, Staten
Island, New York 10314, United States
| | - Tayyaba Hasan
- Wellman
Center for Photomedicine, Massachusetts
General Hospital and Harvard Medical School, 40 Blossom Street, Boston, Massachusetts 02114, United States
- Division
of Health Sciences and Technology, Harvard
University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
66
|
Casula L, Elena Giacomazzo G, Conti L, Fornasier M, Manca B, Schlich M, Sinico C, Rheinberger T, Wurm FR, Giorgi C, Murgia S. Polyphosphoester-stabilized cubosomes encapsulating a Ru(II) complex for the photodynamic treatment of lung adenocarcinoma. J Colloid Interface Sci 2024; 670:234-245. [PMID: 38761576 DOI: 10.1016/j.jcis.2024.05.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
The clinical translation of photosensitizers based on ruthenium(II) polypyridyl complexes (RPCs) in photodynamic therapy of cancer faces several challenges. To address these limitations, we conducted an investigation to assess the potential of a cubosome formulation stabilized in water against coalescence utilizing a polyphosphoester analog of Pluronic F127 as a stabilizer and loaded with newly synthesized RPC-based photosensitizer [Ru(dppn)2(bpy-morph)](PF6)2 (bpy-morph = 2,2'-bipyridine-4,4'-diylbis(morpholinomethanone)), PS-Ru. The photophysical characterization of PS-Ru revealed its robust capacity to induce the formation of singlet oxygen (1O2). Furthermore, the physicochemical analysis of the PS-Ru-loaded cubosomes dispersion demonstrated that the encapsulation of the photosensitizer within the nanoparticles did not disrupt the three-dimensional arrangement of the lipid bilayer. The biological tests showed that PS-Ru-loaded cubosomes exhibited significant phototoxic activity when exposed to the light source, in stark contrast to empty cubosomes and to the same formulation without irradiation. This promising outcome suggests the potential of the formulation in overcoming the drawbacks associated with the clinical use of RPCs in photodynamic therapy for anticancer treatments.
Collapse
Affiliation(s)
- Luca Casula
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, S.P. 8 Km 0.700, 09042 Monserrato, CA, Italy
| | - Gina Elena Giacomazzo
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, FI, Italy
| | - Luca Conti
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, FI, Italy
| | - Marco Fornasier
- Department of Chemistry, Lund University, SE-22100 Lund, Sweden; CSGI, Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, 50019 Sesto Fiorentino, FI, Italy
| | - Benedetto Manca
- Department of Mathematics and Computer Science, University of Cagliari, via Ospedale 72, 09124 Cagliari, CA, Italy
| | - Michele Schlich
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, S.P. 8 Km 0.700, 09042 Monserrato, CA, Italy
| | - Chiara Sinico
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, S.P. 8 Km 0.700, 09042 Monserrato, CA, Italy
| | - Timo Rheinberger
- Sustainable Polymer Chemistry (SPC), Department of Molecules and Materials, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, P.O. Box 217, Enschede 7500 AE, Netherlands
| | - Frederik R Wurm
- Sustainable Polymer Chemistry (SPC), Department of Molecules and Materials, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, P.O. Box 217, Enschede 7500 AE, Netherlands
| | - Claudia Giorgi
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, FI, Italy
| | - Sergio Murgia
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, S.P. 8 Km 0.700, 09042 Monserrato, CA, Italy; CSGI, Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, 50019 Sesto Fiorentino, FI, Italy.
| |
Collapse
|
67
|
Wang J, Zhao S, Yi J, Sun Y, Agrawal M, Oelze ML, Li K, Moore JS, Chen YS. Injectable Mechanophore Nanoparticles for Deep-Tissue Mechanochemical Dynamic Therapy. ACS NANO 2024. [PMID: 39250826 DOI: 10.1021/acsnano.4c04090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Photodynamic therapy (PDT) and sonodynamic therapy (SDT), using nonionizing light and ultrasound to generate reactive oxygen species, offer promising localized treatments for cancers. However, the effectiveness of PDT is hampered by inadequate tissue penetration, and SDT largely relies on pyrolysis and sonoluminescence, which may cause tissue injury and imprecise targeting. To address these issues, we have proposed a mechanochemical dynamic therapy (MDT) that uses free radicals generated from mechanophore-embedded polymers under mechanical stress to produce reactive oxygen species for cancer treatment. Yet, their application in vivo is constrained by the bulk form of the polymer and the need for high ultrasound intensities for activation. In this study, we developed injectable, nanoscale mechanophore particles with enhanced ultrasound sensitivity by leveraging a core-shell structure comprising silica nanoparticles (NPs) whose interfaces are linked to polymer brushes by an azo mechanophore moiety. Upon focused ultrasound (FUS) treatment, this injectable NP generates reactive oxygen species (ROS), demonstrating promising results in both an in vitro 4T1 cell model and an in vivo mouse model of orthotopic breast cancers. This research offers an alternative therapy technique, integrating force-responsive azo mechanophores and FUS under biocompatible conditions.
Collapse
Affiliation(s)
- Jian Wang
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Shensheng Zhao
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Nick Holonyak Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Junxi Yi
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Nick Holonyak Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yunyan Sun
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Megha Agrawal
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Nick Holonyak Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Michael L Oelze
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Nick Holonyak Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - King Li
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Materials Science and Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Jeffrey S Moore
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yun-Sheng Chen
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Nick Holonyak Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
68
|
Morais JAV, Barros PHA, Brigido MDM, Marina CL, Bocca A, Mariano ADLES, Souza PEND, Paiva KLR, Simões MM, Bao SN, Camargo LC, Longo JPF, Morais AAC, Azevedo RBD, Fonseca MJP, Muehlmann LA. Direct and Abscopal Antitumor Responses Elicited by AlPcNE-Mediated Photodynamic Therapy in a Murine Melanoma Model. Pharmaceutics 2024; 16:1177. [PMID: 39339213 PMCID: PMC11435272 DOI: 10.3390/pharmaceutics16091177] [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: 07/12/2024] [Revised: 08/19/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
Melanoma, the most aggressive form of skin cancer, presents a major clinical challenge due to its tendency to metastasize and recalcitrance to traditional therapies. Despite advances in surgery, chemotherapy, and radiotherapy, the outlook for advanced melanoma remains bleak, reinforcing the urgent need for more effective treatments. Photodynamic therapy (PDT) has emerged as a promising alternative, leading to targeted tumor destruction with minimal harm to surrounding tissues. In this study, the direct and abscopal antitumor effects of PDT in a bilateral murine melanoma model were evaluated. Although only one of the two tumors was treated, effects were observed in both. Our findings revealed significant changes in systemic inflammation and alterations in CD4+ and CD8+ T cell populations in treated groups, as evidenced by blood analyses and flow cytometry. High-throughput RNA sequencing (RNA-Seq) further unveiled shifts in gene expression profiles in both treated and untreated tumors. This research sheds light on the novel antitumor and abscopal effects of nanoemulsion of aluminum chloride phthalocyanine (AlPcNE)-mediated PDT in melanoma, highlighting the potential of different PDT protocols to modulate immune responses and to achieve more effective and targeted cancer treatments.
Collapse
Affiliation(s)
- José Athayde Vasconcelos Morais
- Laboratory of Nanoscience and Immunology, Faculty of Ceilandia, University of Brasilia Ceilandia Sul, Brasilia 72220-275, DF, Brazil
- Laboratory of Gene Regulation and Mutagenesis, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Pedro H A Barros
- Laboratory of Molecular Immunology, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Marcelo de Macedo Brigido
- Laboratory of Molecular Immunology, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Clara Luna Marina
- Laboratory of Applied Immunology, Institute of Biology Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Anamelia Bocca
- Laboratory of Applied Immunology, Institute of Biology Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - André de Lima E Silva Mariano
- Laboratory for Softwares and Physics Instrumentation Development, Institute of Physics, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Paulo E N de Souza
- Laboratory for Softwares and Physics Instrumentation Development, Institute of Physics, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Karen L R Paiva
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Marina Mesquita Simões
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Sonia Nair Bao
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Luana C Camargo
- Laboratory of Nanoscience and Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - João P Figueiró Longo
- Laboratory of Nanoscience and Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Amanda Alencar Cabral Morais
- Laboratory of Nanoscience and Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Ricardo B de Azevedo
- Laboratory of Nanoscience and Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Marcio J P Fonseca
- Laboratory of Gene Regulation and Mutagenesis, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Luis A Muehlmann
- Laboratory of Nanoscience and Immunology, Faculty of Ceilandia, University of Brasilia Ceilandia Sul, Brasilia 72220-275, DF, Brazil
| |
Collapse
|
69
|
Talaee O, Faghihi R, Rastegari B, Sina S. Enhanced radio-photodynamic therapy potential of advanced gold-based nanoclusters for breast cancer treatment. Radiol Phys Technol 2024; 17:703-714. [PMID: 39014282 DOI: 10.1007/s12194-024-00824-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/31/2024] [Accepted: 06/23/2024] [Indexed: 07/18/2024]
Abstract
The purpose of current study was to assess the impact of ALA-coated gold nanoclusters (Au NPs) on the combined therapeutic effects of radiotherapy (RT) and photodynamic therapy (PDT) on healthy MCF-10A and MCF-7 breast cancer cells. The Au NPs were covered with ALA using PEG polymer, resulting in the synthesis of Au@ALA NPs. The successful synthesis of the final NPs was confirmed through FTIR, XRD, TEM, and UV-Vis tests. MCF-10A and MCF-7 cell lines were treated with different concentrations of Au@ALA NPs and exposed to irradiation of 2 and 4 Gy (using MV X-ray) and 630 nm laser light irradiation. Cytotoxicity was assessed using a multifaceted approach involving the MTT assay, real-time PCR, and colony forming assay. The findings revealed that the damage inflicted by Au@ALA NPs on cancerous tissue was significantly greater than that on normal tissue. The cytotoxic effects of all experimental groups exhibited a direct correlation with increasing concentrations and radiation doses. The combination of Au@ALA NPs with RT doses of 2 and 4 Gy resulted in a reduction in cell viability by a factor of 1.58 (P = 0.001) and 1.73 (P = 0.004), respectively. Furthermore, the simultaneous intervention of NPs with PDT and RT at doses of 2 and 4 Gy led to a decrease in cell viability by a factor of 2.10 (P = 0.001) and 3.08 (P = 0.001) in turn. Furthermore, the real-time PCR and colonogenic assay results demonstrated that the combined treatment significantly increased phosphorylation of ATM and expression of TP53, indicating an adequate synergistic effect on breast cancer cells. The concurrent application of Au@ALA NPs in RT and PDT successfully enhanced the radiosensitization of breast cancer cells to megavoltage RT and PDT.
Collapse
Affiliation(s)
- Omid Talaee
- Nuclear Engineering Department, Shiraz University, Shiraz, Iran
| | - Reza Faghihi
- Nuclear Engineering Department, Shiraz University, Shiraz, Iran.
- Radiation Research Center, Shiraz University, Shiraz, Iran.
| | - Banafsheh Rastegari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sedigheh Sina
- Nuclear Engineering Department, Shiraz University, Shiraz, Iran
- Radiation Research Center, Shiraz University, Shiraz, Iran
| |
Collapse
|
70
|
Kejík Z, Hajduch J, Abramenko N, Vellieux F, Veselá K, Fialová JL, Petrláková K, Kučnirová K, Kaplánek R, Tatar A, Skaličková M, Masařík M, Babula P, Dytrych P, Hoskovec D, Martásek P, Jakubek M. Cyanine dyes in the mitochondria-targeting photodynamic and photothermal therapy. Commun Chem 2024; 7:180. [PMID: 39138299 PMCID: PMC11322665 DOI: 10.1038/s42004-024-01256-6] [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: 02/08/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024] Open
Abstract
Mitochondrial dysregulation plays a significant role in the carcinogenesis. On the other hand, its destabilization strongly represses the viability and metastatic potential of cancer cells. Photodynamic and photothermal therapies (PDT and PTT) target mitochondria effectively, providing innovative and non-invasive anticancer therapeutic modalities. Cyanine dyes, with strong mitochondrial selectivity, show significant potential in enhancing PDT and PTT. The potential and limitations of cyanine dyes for mitochondrial PDT and PTT are discussed, along with their applications in combination therapies, theranostic techniques, and optimal delivery systems. Additionally, novel approaches for sonodynamic therapy using photoactive cyanine dyes are presented, highlighting advances in cancer treatment.
Collapse
Affiliation(s)
- Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic.
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic.
| | - Jan Hajduch
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Nikita Abramenko
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Frédéric Vellieux
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | | | - Kateřina Petrláková
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00, Brno, Czech Republic
| | - Kateřina Kučnirová
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Ameneh Tatar
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Markéta Skaličková
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Michal Masařík
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Petr Dytrych
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 121 08, Prague, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 121 08, Prague, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic.
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic.
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic.
| |
Collapse
|
71
|
Ajuwon OR, Nsole-Biteghe FA, Ndong JD, Davids LM, Ajiboye BO, Brai B, Bamisaye FA, Falode JA, Odoh IM, Adegbite KI, Adegoke BO, Ntwasa M, Lebelo SL, Ayeleso AO. Nrf2-Mediated Antioxidant Response and Drug Efflux Transporters Upregulation as Possible Mechanisms of Resistance in Photodynamic Therapy of Cancers. Onco Targets Ther 2024; 17:605-627. [PMID: 39131905 PMCID: PMC11313505 DOI: 10.2147/ott.s457749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/08/2024] [Indexed: 08/13/2024] Open
Abstract
Photodynamic therapy (PDT) is a groundbreaking approach involving the induction of cytotoxic reactive oxygen species (ROS) within tumors through visible light activation of photosensitizers (PS) in the presence of molecular oxygen. This innovative therapy has demonstrated success in treating various cancers. While PDT proves highly effective in most solid tumors, there are indications that certain cancers exhibit resistance, and some initially responsive cancers may develop intrinsic or acquired resistance to PDT. The molecular mechanisms underlying this resistance are not fully understood. Recent evidence suggests that, akin to other traditional cancer treatments, the activation of survival pathways, such as the KEAP1/Nrf2 signaling pathway, is emerging as an important mechanism of post-PDT resistance in many cancers. This article explores the dual role of Nrf2, highlighting evidence linking aberrant Nrf2 expression to treatment resistance across a range of cancers. Additionally, it delves into the specific role of Nrf2 in the context of photodynamic therapy for cancers, emphasizing evidence that suggests Nrf2-mediated upregulation of antioxidant responses and induction of drug efflux transporters are potential mechanisms of resistance to PDT in diverse cancer types. Therefore, understanding the specific role(s) of Nrf2 in PDT resistance may pave the way for the development of more effective cancer treatments using PDT.
Collapse
Affiliation(s)
| | | | | | | | | | - Bartholomew Brai
- Department of Biochemistry, Federal University, Oye-Ekiti, Ekiti State, Nigeria
| | | | - John Adeolu Falode
- Department of Biochemistry, Federal University, Oye-Ekiti, Ekiti State, Nigeria
| | - Ikenna Maximillian Odoh
- Department of Biochemistry, Federal University, Oye-Ekiti, Ekiti State, Nigeria
- Medical Center, Federal University, Oye-Ekiti, Ekiti-State, Nigeria
| | - Kabirat Iyabode Adegbite
- Department of Environmental Health Science, College of Basic Medical and Health Sciences, Fountain University, Osogbo, Osun State, Nigeria
| | | | - Monde Ntwasa
- Department of Life and Consumer Sciences, University of South Africa, Florida Park 1709, Roodeport, South Africa
| | - Sogolo Lucky Lebelo
- Department of Life and Consumer Sciences, University of South Africa, Florida Park 1709, Roodeport, South Africa
| | - Ademola Olabode Ayeleso
- Department of Life and Consumer Sciences, University of South Africa, Florida Park 1709, Roodeport, South Africa
- Biochemistry Programme, Bowen University, Iwo, Osun State, Nigeria
| |
Collapse
|
72
|
Burloiu AM, Ozon EA, Musuc AM, Anastasescu M, Socoteanu RP, Atkinson I, Culita DC, Anuta V, Popescu IA, Lupuliasa D, Mihai DP, Gîrd CE, Boscencu R. Porphyrin Photosensitizers into Polysaccharide-Based Biopolymer Hydrogels for Topical Photodynamic Therapy: Physicochemical and Pharmacotechnical Assessments. Gels 2024; 10:499. [PMID: 39195028 DOI: 10.3390/gels10080499] [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: 07/04/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Photodynamic therapy (PDT) is an emerging treatment modality that utilizes light-sensitive compounds, known as photosensitizers, to produce reactive oxygen species (ROS) that can selectively destroy malignant or diseased tissues upon light activation. This study investigates the incorporation of two porphyrin structures, 5-(4-hydroxy-3-methoxyphenyl)-10,15,20-tris-(4-acetoxy-3-methoxyphenyl) porphyrin (P2.2.) and 5,10,15,20-tetrakis-(4-acetoxy-3-methoxyphenyl) porphyrin (P2.1.), into hydroxypropyl cellulose (HPC) hydrogels for potential use in topical photodynamic therapy (PDT). The structural and compositional properties of the resulting hydrogels were characterized using advanced techniques such as Fourier-transform infrared (FTIR) spectroscopy, X-ray diffraction (XRD), thermogravimetric analysis (TGA), atomic force microscopy (AFM), UV-Visible (UV-Vis) spectroscopy, and fluorescence spectroscopy. FTIR spectra revealed a slight shift of the main characteristic absorption bands corresponding to the porphyrins and their interactions with the HPC matrix, indicating successful incorporation and potential hydrogen bonding. XRD patterns revealed the presence of crystalline domains within the HPC matrix, indicating partial crystallization of the porphyrins dispersed within the amorphous polymer structure. TGA results indicated enhanced thermal stability of the HPC-porphyrin gels compared to 10% HPC gel, with additional weight loss stages corresponding to the thermal degradation of the porphyrins. Rheological analysis showed that the gels exhibited pseudoplastic behavior and thixotropic properties, with minimal impact on the flow properties of HPC by P2.1., but notable changes in viscosity and shear stress with P2.2. incorporation, indicating structural modifications. AFM imaging revealed a homogeneous distribution of porphyrins, and UV-Vis and fluorescence spectroscopy confirmed the retention of their photophysical properties. Pharmacotechnical evaluations showed that the hydrogels possessed suitable mechanical properties, optimal pH, high swelling ratios, and excellent spreadability, making them ideal for topical application. These findings suggest that the porphyrin-incorporated HPC hydrogels have significant potential as effective therapeutic agents for topical applications.
Collapse
Affiliation(s)
- Andreea Mihaela Burloiu
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia St., 020956 Bucharest, Romania
| | - Emma Adriana Ozon
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia St., 020956 Bucharest, Romania
| | - Adina Magdalena Musuc
- Institute of Physical Chemistry-Ilie Murgulescu, Romanian Academy, 060021 Bucharest, Romania
| | - Mihai Anastasescu
- Institute of Physical Chemistry-Ilie Murgulescu, Romanian Academy, 060021 Bucharest, Romania
| | - Radu Petre Socoteanu
- Institute of Physical Chemistry-Ilie Murgulescu, Romanian Academy, 060021 Bucharest, Romania
| | - Irina Atkinson
- Institute of Physical Chemistry-Ilie Murgulescu, Romanian Academy, 060021 Bucharest, Romania
| | - Daniela C Culita
- Institute of Physical Chemistry-Ilie Murgulescu, Romanian Academy, 060021 Bucharest, Romania
| | - Valentina Anuta
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia St., 020956 Bucharest, Romania
| | - Ioana Andreea Popescu
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia St., 020956 Bucharest, Romania
| | - Dumitru Lupuliasa
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia St., 020956 Bucharest, Romania
| | - Dragoș Paul Mihai
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia St., 020956 Bucharest, Romania
| | - Cerasela Elena Gîrd
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia St., 020956 Bucharest, Romania
| | - Rica Boscencu
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia St., 020956 Bucharest, Romania
| |
Collapse
|
73
|
An J, Lv KP, Chau CV, Lim JH, Parida R, Huang X, Debnath S, Xu Y, Zheng S, Sedgwick AC, Lee JY, Luo D, Liu Q, Sessler JL, Kim JS. Lutetium Texaphyrin-Celecoxib Conjugate as a Potential Immuno-Photodynamic Therapy Agent. J Am Chem Soc 2024; 146:19434-19448. [PMID: 38959476 PMCID: PMC12005638 DOI: 10.1021/jacs.4c05978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Immuno-photodynamic therapy (IPDT) has emerged as a new modality for cancer treatment. Novel photosensitizers can help achieve the promise inherent in IPDT, namely, the complete eradication of a tumor without recurrence. We report here a small molecule photosensitizer conjugate, LuCXB. This IPDT agent integrates a celecoxib (cyclooxygenase-2 inhibitor) moiety with a near-infrared absorbing lutetium texaphyrin photocatalytic core. In aqueous environments, the two components of LuCXB are self-associated through inferred donor-acceptor interactions. A consequence of this intramolecular association is that upon photoirradiation with 730 nm light, LuCXB produces superoxide radicals (O2-•) via a type I photodynamic pathway; this provides a first line of defense against the tumor while promoting IPDT. For in vivo therapeutic applications, we prepared a CD133-targeting, aptamer-functionalized exosome-based nanophotosensitizer (Ex-apt@LuCXB) designed to target cancer stem cells. Ex-apt@LuCXB was found to display good photosensitivity, acceptable biocompatibility, and robust tumor targetability. Under conditions of photoirradiation, Ex-apt@LuCXB acts to amplify IPDT while exerting a significant antitumor effect in both liver and breast cancer mouse models. The observed therapeutic effects are attributed to a synergistic mechanism that combines antiangiogenesis and photoinduced cancer immunotherapy.
Collapse
Affiliation(s)
- Jusung An
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Kong-Peng Lv
- Laboratory Medicine Center, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen 518000 Guangdong, China; Department of Interventional Radiology, Shenzhen People’s Hospital, (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020 Guangdong, China
| | - Calvin V. Chau
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jong Hyeon Lim
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Korea
| | - Rakesh Parida
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Korea
| | - Xin Huang
- Laboratory Medicine Center, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen 518000 Guangdong, China
| | | | - Yunjie Xu
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Siqi Zheng
- Department of Interventional Radiology, Shenzhen People’s Hospital, (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020 Guangdong, China
| | - Adam C. Sedgwick
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jin Yong Lee
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Korea
| | - Dixian Luo
- Laboratory Medicine Center, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen 518000 Guangdong, China
| | - Quan Liu
- Laboratory Medicine Center, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen 518000 Guangdong, China
| | - Jonathan L. Sessler
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Korea
| |
Collapse
|
74
|
Liu KT, Wang PW, Hsieh HY, Pan HC, Chin HJ, Lin CW, Huang YJ, Liao YC, Tsai YC, Liu SR, Su IC, Song YF, Yin GC, Wu KC, Chuang EY, Fan YJR, Yu J. Site-specific thrombus formation: advancements in photothrombosis-on-a-chip technology. LAB ON A CHIP 2024; 24:3422-3433. [PMID: 38860416 DOI: 10.1039/d4lc00216d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Thrombosis, characterized by blood clot formation within vessels, poses a significant medical challenge. Despite extensive research, the development of effective thrombosis therapies is hindered by substantial costs, lengthy development times, and high failure rates in medication commercialization. Conventional pre-clinical models often oversimplify cardiovascular disease, leading to a disparity between experimental results and human physiological responses. In response, we have engineered a photothrombosis-on-a-chip system. This microfluidic model integrates human endothelium, human whole blood, and blood flow dynamics and employs the photothrombotic method. It enables precise, site-specific thrombus induction through controlled laser irradiation, effectively mimicking both normal and thrombotic physiological conditions on a single chip. Additionally, the system allows for the fine-tuning of thrombus occlusion levels via laser parameter adjustments, offering a flexible thrombus model with varying degrees of obstruction. Additionally, the formation and progression of thrombosis noted on the chip closely resemble the thrombotic conditions observed in mice in previous studies. In the experiments, we perfused recalcified whole blood with Rose Bengal into an endothelialized microchannel and initiated photothrombosis using green laser irradiation. Various imaging methods verified the model's ability to precisely control thrombus formation and occlusion levels. The effectiveness of clinical drugs, including heparin and rt-PA, was assessed, confirming the chip's potential in drug screening applications. In summary, the photothrombosis-on-a-chip system significantly advances human thrombosis modeling. Its precise control over thrombus formation, flexibility in the thrombus severity levels, and capability to simulate dual physiological states on a single platform make it an invaluable tool for targeted drug testing, furthering the development of organ-on-a-chip drug screening techniques.
Collapse
Affiliation(s)
- Kuan-Ting Liu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| | - Pai-Wen Wang
- Institute of Applied Mechanics, National Taiwan University, Taipei 10617, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Han-Yun Hsieh
- Department of Biochemical and Molecular Medical Science, National Dong Hwa University, Hualien 97401, Taiwan
| | - Han-Chi Pan
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei 115021, Taiwan
| | - Hsian-Jean Chin
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei 115021, Taiwan
| | - Che-Wei Lin
- School of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yu-Jen Huang
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| | - Yung-Chieh Liao
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| | - Ya-Chun Tsai
- Institute of Applied Mechanics, National Taiwan University, Taipei 10617, Taiwan
| | - Shang-Ru Liu
- Institute of Applied Mechanics, National Taiwan University, Taipei 10617, Taiwan
| | - I-Chang Su
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Neurosurgery, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, 23561, Taiwan
| | - Yen-Fang Song
- National Synchrotron Radiation Research Center, Hsinchu 300092, Taiwan
| | - Gung-Chian Yin
- National Synchrotron Radiation Research Center, Hsinchu 300092, Taiwan
| | - Kuang-Chong Wu
- Institute of Applied Mechanics, National Taiwan University, Taipei 10617, Taiwan
| | - Er-Yuan Chuang
- School of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yu-Jui Ray Fan
- School of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| | - Jiashing Yu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
75
|
Mohanty S, Desai VM, Jain R, Agrawal M, Dubey SK, Singhvi G. Unveiling the potential of photodynamic therapy with nanocarriers as a compelling therapeutic approach for skin cancer treatment: current explorations and insights. RSC Adv 2024; 14:21915-21937. [PMID: 38989245 PMCID: PMC11234503 DOI: 10.1039/d4ra02564d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
Skin carcinoma is one of the most prevalent types of carcinomas. Due to high incidence of side effects in conventional therapies (radiotherapy and chemotherapy), photodynamic therapy (PDT) has gained huge attention as an alternate treatment strategy. PDT involves the administration of photosensitizers (PS) to carcinoma cells which produce reactive oxygen species (ROS) on irradiation by specific wavelengths of light that result in cancer cells' death via apoptosis, autophagy, or necrosis. Topical delivery of PS to the skin cancer cells at the required concentration is a challenge due to the compounds' innate physicochemical characteristics. Nanocarriers have been observed to improve skin permeability and enhance the therapeutic efficiency of PDT. Polymeric nanoparticles (NPs), metallic NPs, and lipid nanocarriers have been reported to carry PS successfully with minimal side effects and high effectiveness in both melanoma and non-melanoma skin cancers. Advanced carriers such as quantum dots, microneedles, and cubosomes have also been addressed with reported studies to show their scope of use in PDT-assisted skin cancer treatment. In this review, nanocarrier-aided PDT in skin cancer therapies has been discussed with clinical trials and patents. Additionally, novel nanocarriers that are being investigated in PDT are also covered with their future prospects in skin carcinoma treatment.
Collapse
Affiliation(s)
- Shambo Mohanty
- Industrial Research Laboratory, Department of Pharmacy, FD-III, Birla Institute of Technology and Science, Pilani (BITS-PILANI) Pilani Campus, Vidya Vihar Pilani Rajasthan 333031 India
| | - Vaibhavi Meghraj Desai
- Industrial Research Laboratory, Department of Pharmacy, FD-III, Birla Institute of Technology and Science, Pilani (BITS-PILANI) Pilani Campus, Vidya Vihar Pilani Rajasthan 333031 India
| | - Rupesh Jain
- Industrial Research Laboratory, Department of Pharmacy, FD-III, Birla Institute of Technology and Science, Pilani (BITS-PILANI) Pilani Campus, Vidya Vihar Pilani Rajasthan 333031 India
| | - Mukta Agrawal
- School of Pharmacy & Technology Management, NMIMS Hyderabad India
| | | | - Gautam Singhvi
- Industrial Research Laboratory, Department of Pharmacy, FD-III, Birla Institute of Technology and Science, Pilani (BITS-PILANI) Pilani Campus, Vidya Vihar Pilani Rajasthan 333031 India
| |
Collapse
|
76
|
Tian S, Nie Q, Chen H, Liang L, Hu H, Tang S, Yang J, Liu Y, Yin H. Synthesis, characterization and irradiation enhances anticancer activity of liposome-loaded iridium(III) complexes. J Inorg Biochem 2024; 256:112549. [PMID: 38579631 DOI: 10.1016/j.jinorgbio.2024.112549] [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: 01/24/2024] [Revised: 03/18/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024]
Abstract
Herein, we synthesized and characterized two novel iridium (III) complexes: [Ir(bzq)2(PPD)](PF6) (4a, with bzq = deprotonated benzo[h]quinoline and PPD = pteridino[6,7-f][1,10]phenanthroline-11,13-diamine) and [Ir(piq)2(PPD)](PF6) (4b, with piq = deprotonated 1-phenylisoquinoline). The anticancer efficacy of these complexes, 4a and 4b, was investigated using 3-(4,5-dimethylthiazole)-2,5-diphenltetraazolium bromide (MTT). Complex 4a exhibited no cytotoxic activity, while 4b demonstrated moderate efficacy against SGC-7901, A549, and HepG2 cancer cells. To enhance their anticancer potential, we explored two strategies: (I) light irradiation and (II) encapsulation of the complexes in liposomes, resulting in the formation of 4alip and 4blip. Both strategies significantly increased the ability of 4a, 4b to kill cancer cells. The cellular studies indicated that both the free complexes 4a, 4b and their liposomal forms 4alip and 4blip effectively inhibited cell proliferation. The cell cycle arrest analysis uncovered 4alip and 4blip arresting cell growth in the S period. Additionally, we investigated apoptosis and ferroptosis pathways, observing an increase in malondialdehyde (MDA) levels, a reduction of glutathione (GSH), a down-regulation of GPX4 (glutathione peroxidase) expression, and lipid peroxidation. The effects on mitochondrial membrane potential and intracellular Ca2+ concentrations were also examined, revealing that both light-activated and liposomal forms of 4alip and 4blip caused a decline in mitochondrial membrane potential and an enhancement in intracellular Ca2+ levels. In conclusion, these complexes and them encapsulated liposomes induce cell death through apoptosis and ferroptosis.
Collapse
Affiliation(s)
- Shuang Tian
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qianying Nie
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Haomin Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lijuan Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huiyan Hu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shuanghui Tang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiawan Yang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Hui Yin
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
77
|
Shirke AA, Walker E, Chavali S, Ramamurthy G, Zhang L, Panigrahi A, Basilion JP, Wang X. A Synergistic Strategy Combining Chemotherapy and Photodynamic Therapy to Eradicate Prostate Cancer. Int J Mol Sci 2024; 25:7086. [PMID: 39000194 PMCID: PMC11241360 DOI: 10.3390/ijms25137086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Prostate cancer is the most prevalent cancer among men in the United States and is a leading cause of cancer-related death. Prostate specific membrane antigen (PSMA) has been established as a biomarker for prostate cancer diagnosis and treatment. This study aimed to develop a novel theranostic agent, PSMA-1-MMAE-Pc413, which integrates a PSMA-targeting ligand, the photosensitizer Pc413, and the microtubular inhibitor monomethyl auristatin E (MMAE) for synergistic therapeutic efficacy. In vitro uptake studies revealed that PSMA-1-MMAE-Pc413 demonstrated selective and specific uptake in PSMA-positive PC3pip cells but not in PSMA-negative PC3flu cells, with the uptake in PC3pip cells being approximately three times higher. In vitro cytotoxicity assays showed that, when exposed to light, PSMA-1-MMAE-Pc413 had a synergistic effect, leading to significantly greater cytotoxicity in PSMA-positive cells (IC50 = 2.2 nM) compared to PSMA-1-Pc413 with light irradiation (IC50 = 164.9 nM) or PSMA-1-MMAE-Pc413 without light irradiation (IC50 = 12.6 nM). In vivo imaging studies further demonstrated the selective uptake of PSMA-1-MMAE-Pc413 in PC3pip tumors. In in vivo studies, PSMA-1-MMAE-Pc413 dramatically improves the therapeutic outcome for prostate cancer by providing a synergistic effect that surpasses the efficacy of each treatment modality alone in PC3pip tumors. These findings suggest that PSMA-1-MMAE-Pc413 has strong potential for clinical application in improving prostate cancer treatment.
Collapse
Affiliation(s)
- Aditi A. Shirke
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106, USA; (A.A.S.); (E.W.)
| | - Ethan Walker
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106, USA; (A.A.S.); (E.W.)
| | - Sriprada Chavali
- Department of Biochemistry, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106, USA;
| | - Gopalakrishnan Ramamurthy
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106, USA; (G.R.); (L.Z.); (A.P.)
| | - Lifang Zhang
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106, USA; (G.R.); (L.Z.); (A.P.)
| | - Abhiram Panigrahi
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106, USA; (G.R.); (L.Z.); (A.P.)
| | - James P. Basilion
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106, USA; (A.A.S.); (E.W.)
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106, USA; (G.R.); (L.Z.); (A.P.)
| | - Xinning Wang
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106, USA; (A.A.S.); (E.W.)
| |
Collapse
|
78
|
Wu L, He C, Zhao T, Li T, Xu H, Wen J, Xu X, Gao L. Diagnosis and treatment status of inoperable locally advanced breast cancer and the application value of inorganic nanomaterials. J Nanobiotechnology 2024; 22:366. [PMID: 38918821 PMCID: PMC11197354 DOI: 10.1186/s12951-024-02644-9] [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: 03/26/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024] Open
Abstract
Locally advanced breast cancer (LABC) is a heterogeneous group of breast cancer that accounts for 10-30% of breast cancer cases. Despite the ongoing development of current treatment methods, LABC remains a severe and complex public health concern around the world, thus prompting the urgent requirement for innovative diagnosis and treatment strategies. The primary treatment challenges are inoperable clinical status and ineffective local control methods. With the rapid advancement of nanotechnology, inorganic nanoparticles (INPs) exhibit a potential application prospect in diagnosing and treating breast cancer. Due to the unique inherent characteristics of INPs, different functions can be performed via appropriate modifications and constructions, thus making them suitable for different imaging technology strategies and treatment schemes. INPs can improve the efficacy of conventional local radiotherapy treatment. In the face of inoperable LABC, INPs have proposed new local therapeutic methods and fostered the evolution of novel strategies such as photothermal and photodynamic therapy, magnetothermal therapy, sonodynamic therapy, and multifunctional inorganic nanoplatform. This article reviews the advances of INPs in local accurate imaging and breast cancer treatment and offers insights to overcome the existing clinical difficulties in LABC management.
Collapse
Affiliation(s)
- Linxuan Wu
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China
| | - Chuan He
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Tingting Zhao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Tianqi Li
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China
| | - Hefeng Xu
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China
| | - Jian Wen
- Department of Breast Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| | - Xiaoqian Xu
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China.
| | - Lin Gao
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| |
Collapse
|
79
|
Cui X, Yuan H, Chen X, Meng Q, Zhang C. Newly Designed Quasi-intrinsic Photosensitizers for Fluorescence Image-Guided Two-Photon Photodynamic Therapy with Type I/II Photoreactions. J Med Chem 2024; 67:8902-8912. [PMID: 38815214 DOI: 10.1021/acs.jmedchem.4c00191] [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: 06/01/2024]
Abstract
In this work, a set of quasi-intrinsic photosensitizers are theoretically proposed based on the 2-amino-8-(1'-β-d-2'-deoxyribofuranosyl)-imidazo[1,2-α]-1,3,5-triazin-4(8H)-one (P), which could pair with the 6-amino-5-nitro-3-(1'-β-d-2'-deoxyribofuranosyl)-2(1H)-pyridone (Z) and keep the essential structural characters of nucleic acid. It is revealed that the ring expansion and electron-donating/electron-withdrawing substitution bring enhanced two-photon absorption and bright photoluminescence of these monomers, thereby facilitating the selective excitation and tumor localization through fluorescence imaging. However, instead of undergoing radiative transition (S1 → S0), the base pairing induced fluorescence quenching and rapid intersystem crossing (S1 → Tn) are observed and characterized by the reduced singlet-triplet energy gaps and large spin-orbit coupling values. To ensure the phototherapeutic properties of the considered base pairs in long-lived T1 state, we examined the vertical electron affinity as well as vertical ionization potential for production of superoxide anions via Type I photoreaction, and their required T1 energy (0.98 eV) to generate singlet oxygen 1O2 via Type II mechanism.
Collapse
Affiliation(s)
- Xixi Cui
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, P. R. China
| | - Hongxiu Yuan
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, P. R. China
| | - Xiaolin Chen
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, P. R. China
| | - Qingtian Meng
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, P. R. China
| | - Changzhe Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan 250358, P. R. China
| |
Collapse
|
80
|
Zhen W, Kang DW, Fan Y, Wang Z, Germanas T, Nash GT, Shen Q, Leech R, Li J, Engel GS, Weichselbaum RR, Lin W. Simultaneous Protonation and Metalation of a Porphyrin Covalent Organic Framework Enhance Photodynamic Therapy. J Am Chem Soc 2024. [PMID: 38837955 DOI: 10.1021/jacs.4c03519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Covalent organic frameworks (COFs) have been explored for photodynamic therapy (PDT) of cancer, but their antitumor efficacy is limited by excited state quenching and low reactive oxygen species generation efficiency. Herein, we report a simultaneous protonation and metalation strategy to significantly enhance the PDT efficacy of a nanoscale two-dimensional imine-linked porphyrin-COF. The neutral and unmetalated porphyrin-COF (Ptp) and the protonated and metalated porphyrin-COF (Ptp-Fe) were synthesized via imine condensation between 5,10,15,20-tetrakis(4-aminophenyl)porphyrin and terephthalaldehyde in the absence and presence of ferric chloride, respectively. The presence of ferric chloride generated both doubly protonated and Fe3+-coordinated porphyrin units, which red-shifted and increased the Q-band absorption and disrupted exciton migration to prevent excited state quenching, respectively. Under light irradiation, rapid energy transfer from protonated porphyrins to Fe3+-coordinated porphyrins in Ptp-Fe enabled 1O2 and hydroxyl radical generation via type II and type I PDT processes. Ptp-Fe also catalyzed the conversion of hydrogen peroxide to hydroxy radical through a photoenhanced Fenton-like reaction under slightly acidic conditions and light illumination. As a result, Ptp-Fe-mediated PDT exhibited much higher cytotoxicity than Ptp-mediated PDT on CT26 and 4T1 cancer cells. Ptp-Fe-mediated PDT afforded potent antitumor efficacy in subcutaneous CT26 murine colon cancer and orthotopic 4T1 murine triple-negative breast tumors and prevented metastasis of 4T1 breast cancer to the lungs. This work underscores the role of fine-tuning the molecular structures of COFs in significantly enhancing their PDT efficacy.
Collapse
Affiliation(s)
- Wenyao Zhen
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637, United States
| | - Dong Won Kang
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
- Department of Chemistry and Chemical Engineering, Inha University, 100 Inha-Ro, Michuhol-Gu, Incheon, 22212, Republic of Korea
| | - Yingjie Fan
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Zitong Wang
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Tomas Germanas
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Geoffrey T Nash
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Qijie Shen
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Rachel Leech
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Jinhong Li
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Gregory S Engel
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637, United States
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
81
|
Dos Santos MIB, Godoi BH, Da Silva NS, Oliveira LD, de Paula Ramos L, Cintra RC, Pacheco-Soares C. Modulation of heat shock protein expression and cytokine levels in MCF-7 cells through photodynamic therapy. Lasers Med Sci 2024; 39:135. [PMID: 38787412 DOI: 10.1007/s10103-024-04092-1] [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: 02/15/2024] [Accepted: 05/18/2024] [Indexed: 05/25/2024]
Abstract
In this study, we assess the impact of photodynamic therapy (PDT) using aluminum phthalocyanine tetrasulfonate (AlPcS4) on the viability and cellular stress responses of MCF-7 breast cancer cells. Specifically, we investigate changes in cell viability, cytokine production, and the expression of stress-related genes. Experimental groups included control cells, those treated with AlPcS4 only, light-emitting diode (LED) only, and combined PDT. To evaluate these effects on cell viability, cytokine production, and the expression of stress-related genes, techniques such as 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay, enzyme-linked immunosorbent assays (ELISA), and real-time quantitative PCR (RT‒qPCR) were employed. Our findings reveal how PDT with AlPcS4 modulates mitochondrial activity and cytokine responses, shedding light on the cellular pathways essential for cell survival and stress adaptation. This work enhances our understanding of PDT's therapeutic potential and mechanisms in treating breast cancer.
Collapse
Affiliation(s)
| | - Bruno Henrique Godoi
- Universidade Do Vale Do Paraíba, Av Shishima Hifumi 2911, Urbanova, São José dos Campos, SP, 12244-000, Brazil
| | - Newton Soares Da Silva
- Universidade Estadual Paulista Júlio de Mesquita Filho, Av. Eng. Francisco José Longo, 777 Jardim São Dimas, São José dos Campos, SP, 12245-000, Brazil
- , São José Dos Campos, Brazil
| | - Luciane Dias Oliveira
- Universidade Estadual Paulista Júlio de Mesquita Filho, Av. Eng. Francisco José Longo, 777 Jardim São Dimas, São José dos Campos, SP, 12245-000, Brazil
- , São José Dos Campos, Brazil
| | - Lucas de Paula Ramos
- Universidade Estadual Paulista Júlio de Mesquita Filho, Av. Eng. Francisco José Longo, 777 Jardim São Dimas, São José dos Campos, SP, 12245-000, Brazil
- , São José Dos Campos, Brazil
- University Claude Bernard Lyon, Laboratory Health Systemic Process - P2S, UR4129, Faculty of Medicine Laennec, rue Guillaume Paradin, 69008, Lyon, France
| | - Ricardo Cesar Cintra
- Universidade de São Paulo- R. da Reitoria, 374 Cidade Universitária, Butantã, São Paulo, SP, 05508-220, Brazil
| | - Cristina Pacheco-Soares
- Universidade Do Vale Do Paraíba, Av Shishima Hifumi 2911, Urbanova, São José dos Campos, SP, 12244-000, Brazil.
| |
Collapse
|
82
|
Liu J, Sun B, Li W, Kim HJ, Gan SU, Ho JS, Rahmat JNB, Zhang Y. Wireless sequential dual light delivery for programmed PDT in vivo. LIGHT, SCIENCE & APPLICATIONS 2024; 13:113. [PMID: 38744817 PMCID: PMC11094163 DOI: 10.1038/s41377-024-01437-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/05/2024] [Accepted: 03/21/2024] [Indexed: 05/16/2024]
Abstract
Using photodynamic therapy (PDT) to treat deep-seated cancers is limited due to inefficient delivery of photosensitizers and low tissue penetration of light. Polymeric nanocarriers are widely used for photosensitizer delivery, while the self-quenching of the encapsulated photosensitizers would impair the PDT efficacy. Furthermore, the generated short-lived reactive oxygen spieces (ROS) can hardly diffuse out of nanocarriers, resulting in low PDT efficacy. Therefore, a smart nanocarrier system which can be degraded by light, followed by photosensitizer activation can potentially overcome these limitations and enhance the PDT efficacy. A light-sensitive polymer nanocarrier encapsulating photosensitizer (RB-M) was synthesized. An implantable wireless dual wavelength microLED device which delivers the two light wavelengths sequentially was developed to programmatically control the release and activation of the loaded photosensitizer. Two transmitter coils with matching resonant frequencies allow activation of the connected LEDs to emit different wavelengths independently. Optimal irradiation time, dose, and RB-M concentration were determined using an agent-based digital simulation method. In vitro and in vivo validation experiments in an orthotopic rat liver hepatocellular carcinoma disease model confirmed that the nanocarrier rupture and sequential low dose light irradiation strategy resulted in successful PDT at reduced photosensitizer and irradiation dose, which is a clinically significant event that enhances treatment safety.
Collapse
Affiliation(s)
- Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Bowen Sun
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117585, Singapore
| | - Wenkai Li
- Department of Mechanical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Han-Joon Kim
- Department of Electrical and Computer Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, 39253, Republic of Korea
| | - Shu Uin Gan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - John S Ho
- Department of Electrical and Computer Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore, 117456, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, 119276, Singapore
| | - Juwita Norasmara Bte Rahmat
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117585, Singapore.
| | - Yong Zhang
- Department of Biomedical Engineering, College of Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China.
| |
Collapse
|
83
|
Spadin FS, Gergely LP, Kämpfer T, Frenz M, Vermathen M. Fluorescence lifetime imaging and phasor analysis of intracellular porphyrinic photosensitizers applied with different polymeric formulations. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 254:112904. [PMID: 38579534 DOI: 10.1016/j.jphotobiol.2024.112904] [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: 12/09/2023] [Revised: 03/06/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
The fluorescence lifetime of a porphyrinic photosensitizer (PS) is an important parameter to assess the aggregation state of the PS even in complex biological environments. Aggregation-induced quenching of the PS can significantly reduce the yield of singlet oxygen generation and thus its efficiency as a medical drug in photodynamic therapy (PDT) of diseased tissues. Hydrophobicity and the tendency to form aggregates pose challenges on the development of efficient PSs and often require carrier systems. A systematic study was performed to probe the impact of PS structure and encapsulation into polymeric carriers on the fluorescence lifetime in solution and in the intracellular environment. Five different porphyrinic PSs including chlorin e6 (Ce6) derivatives and tetrakis(m-hydroxyphenyl)-porphyrin and -chlorin were studied in free form and combined with polyvinylpyrrolidone (PVP) or micelles composed of triblock-copolymers or Cremophor. Following incubation of HeLa cells with these systems, fluorescence lifetime imaging combined with phasor analysis and image segmentation was applied to study the lifetime distribution in the intracellular surrounding. The data suggest that for free PSs, the structure-dependent cell uptake pathways determine their state and emission lifetimes. PS localization in the plasma membrane yielded mostly monomers with long fluorescence lifetimes whereas the endocytic pathway with subsequent lysosomal deposition adds a short-lived component for hydrophilic anionic PSs. Prolonged incubation times led to increasing contributions from short-lived components that derive from aggregates mainly localized in the cytoplasm. Encapsulation of PSs into polymeric carriers led to monomerization and mostly fluorescence emission decays with long fluorescence lifetimes in solution. However, the efficiency depended on the binding strength that was most pronounced for PVP. In the cellular environment, PVP was able to maintain monomeric long-lived species over prolonged incubation times. This was most pronounced for Ce6 derivatives with a logP value around 4.5. Micellar encapsulation led to faster release of the PSs resulting in multiple components with long and short fluorescence lifetimes. The hydrophilic hardly aggregating PS exhibited a mostly stable invariant lifetime distribution over time with both carriers. The presented data are expected to contribute to optimized PDT treatment protocols and improved PS-carrier design for preventing intracellular fluorescence quenching. In conclusion, amphiphilic and concurrent hydrophobic PSs with high membrane affinity as well as strong binding to the carrier have best prospects to maintain their photophysical properties in vivo and serve thus as efficient photodynamic diagnosis and PDT drugs.
Collapse
Affiliation(s)
- Florentin S Spadin
- Institute of Applied Physics, University of Bern, Sidlerstrasse 5, 3012 Bern, Switzerland
| | - Lea P Gergely
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, Freiestrasse 3, 3012 Bern, Switzerland
| | - Tobias Kämpfer
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, Freiestrasse 3, 3012 Bern, Switzerland
| | - Martin Frenz
- Institute of Applied Physics, University of Bern, Sidlerstrasse 5, 3012 Bern, Switzerland.
| | - Martina Vermathen
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, Freiestrasse 3, 3012 Bern, Switzerland.
| |
Collapse
|
84
|
Lozano-Rosas R, Ramos-Garcia R, Salazar-Morales MF, Robles-Águila MJ, Spezzia-Mazzocco T. Evaluation of antifungal activity of visible light-activated doped TiO 2 nanoparticles. Photochem Photobiol Sci 2024; 23:823-837. [PMID: 38568410 DOI: 10.1007/s43630-024-00557-y] [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: 09/20/2023] [Accepted: 03/04/2024] [Indexed: 06/11/2024]
Abstract
Titanium dioxide (TiO2) is a well-known material for its biomedical applications, among which its implementation as a photosensitizer in photodynamic therapy has attracted considerable interest due to its photocatalytic properties, biocompatibility, high chemical stability, and low toxicity. However, the photoactivation of TiO2 requires ultraviolet light, which may lead to cell mutation and consequently cancer. To address these challenges, recent research has focused on the incorporation of metal dopants into the TiO2 lattice to shift the band gap to lower energies by introducing allowed energy states within the band gap, thus ensuring the harnessing of visible light. This study presents the synthesis, characterization, and application of TiO2 nanoparticles (NPs) in their undoped, doped, and co-doped forms for antimicrobial photodynamic therapy (APDT) against Candida albicans. Blue light with a wavelength of 450 nm was used, with doses ranging from 20 to 60 J/cm2 and an NP concentration of 500 µg/ml. It was observed that doping TiO2 with Cu, Fe, Ag ions, and co-doping Cu:Fe into the TiO2 nanostructure enhanced the visible light photoactivity of TiO2 NPs. Experimental studies were done to investigate the effects of different ions doped into the TiO2 crystal lattice on their structural, optical, morphological, and chemical composition for APDT applications. In particular, Ag-doped TiO2 emerged as the best candidate, achieving 90-100% eradication of C. albicans.
Collapse
Affiliation(s)
- Ricardo Lozano-Rosas
- Instituto Nacional de Astrofísica, Óptica y Electrónica, Departamento de Óptica, Luis Enrique Erro #1 Sta María Tonantzintla, 72840, Puebla, Mexico
| | - Rubén Ramos-Garcia
- Instituto Nacional de Astrofísica, Óptica y Electrónica, Departamento de Óptica, Luis Enrique Erro #1 Sta María Tonantzintla, 72840, Puebla, Mexico
| | - Mayra F Salazar-Morales
- Instituto Nacional de Astrofísica, Óptica y Electrónica, Departamento de Óptica, Luis Enrique Erro #1 Sta María Tonantzintla, 72840, Puebla, Mexico
| | - María Josefina Robles-Águila
- Centro de Investigación en Dispositivos Semiconductores, Benemérita Universidad Autónoma de Puebla, Instituto de Ciencias, Edificio 105 C, Boulevard 14 Sur y Av. San Claudio, Col. San Manuel, C. P. 72570, Puebla, Puebla, Mexico
| | - Teresita Spezzia-Mazzocco
- Instituto Nacional de Astrofísica, Óptica y Electrónica, Departamento de Óptica, Luis Enrique Erro #1 Sta María Tonantzintla, 72840, Puebla, Mexico.
| |
Collapse
|
85
|
Mesquita B, Singh A, Prats Masdeu C, Lokhorst N, Hebels ER, van Steenbergen M, Mastrobattista E, Heger M, van Nostrum CF, Oliveira S. Nanobody-mediated targeting of zinc phthalocyanine with polymer micelles as nanocarriers. Int J Pharm 2024; 655:124004. [PMID: 38492899 DOI: 10.1016/j.ijpharm.2024.124004] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 03/18/2024]
Abstract
Photodynamic therapy (PDT) is a suitable alternative to currently employed cancer treatments. However, the hydrophobicity of most photosensitizers (e.g., zinc phthalocyanine (ZnPC)) leads to their aggregation in blood. Moreover, non-specific accumulation in skin and low clearance rate of ZnPC leads to long-lasting skin photosensitization, forcing patients with a short life expectancy to remain indoors. Consequently, the clinical implementation of these photosensitizers is limited. Here, benzyl-poly(ε-caprolactone)-b-poly(ethylene glycol) micelles encapsulating ZnPC (ZnPC-M) were investigated to increase the solubility of ZnPC and its specificity towards cancers cells. Asymmetric flow field-flow fractionation was used to characterize micelles with different ZnPC-to-polymer ratios and their stability in human plasma. The ZnPC-M with the lowest payload (0.2 and 0.4% ZnPC w/w) were the most stable in plasma, exhibiting minimal ZnPC transfer to lipoproteins, and induced the highest phototoxicity in three cancer cell lines. Nanobodies (Nbs) with binding specificity towards hepatocyte growth factor receptor (MET) or epidermal growth factor receptor (EGFR) were conjugated to ZnPC-M to facilitate cell targeting and internalization. MET- and EGFR-targeting micelles enhanced the association and the phototoxicity in cells expressing the target receptor. Altogether, these results indicate that ZnPC-M decorated with Nbs targeting overexpressed proteins on cancer cells may provide a better alternative to currently approved formulations.
Collapse
Affiliation(s)
- Bárbara Mesquita
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Arunika Singh
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Cèlia Prats Masdeu
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Nienke Lokhorst
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Erik R Hebels
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Mies van Steenbergen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Michal Heger
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, Jiaxing University, College of Medicine, Jiaxing, Zhejiang, PR China; Membrane Biochemistry and Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Utrecht, The Netherlands
| | - Cornelus F van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| | - Sabrina Oliveira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
86
|
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
|
87
|
Luppi BT, Primrose WL, Hudson ZM. Polymer Dots with Delayed Fluorescence and Tunable Cellular Uptake for Photodynamic Therapy and Time-Gated Imaging. Angew Chem Int Ed Engl 2024; 63:e202400712. [PMID: 38439710 DOI: 10.1002/anie.202400712] [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: 01/11/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/06/2024]
Abstract
By combining bioimaging and photodynamic therapy (PDT), it is possible to treat cancer through a theranostic approach with targeted action for minimum invasiveness and side effects. Thermally activated delayed fluorescence (TADF) probes have gained recent interest in theranostics due to their ability to generate singlet oxygen (1O2) while providing delayed emission that can be used in time-gated imaging. However, it is still challenging to design systems that simultaneously show (1) high contrast for imaging, (2) low dark toxicity but high phototoxicity and (3) tunable biological uptake. Here, we circumvent shortcomings of TADF systems by designing block copolymers and their corresponding semiconducting polymer dots (Pdots) that encapsulate a TADF dye in the core and expose an additional boron-dipyrromethene (BODIPY) oxygen sensitizer in the corona. This architecture provides orange-red luminescent particles (ΦPL up to 18 %) that can efficiently promote PDT (1O2 QY=42 %) of HeLa cells with very low photosensitizer loading (IC50 ~0.05-0.13 μg/mL after 30 min). Additionally, we design Pdots with tunable cellular uptake but similar PDT efficiencies using either polyethylene glycol or guanidinium-based coronas. Finally, we demonstrate that these Pdots can be used for time-gated imaging to effectively filter out background fluorescence from biological samples and improve image contrast.
Collapse
Affiliation(s)
- Bruno T Luppi
- Department of Chemistry, The University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, V6T 1Z1, Canada
| | - William L Primrose
- Department of Chemistry, The University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, V6T 1Z1, Canada
| | - Zachary M Hudson
- Department of Chemistry, The University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, V6T 1Z1, Canada
| |
Collapse
|
88
|
Liang J, Ran X, Liu Y, Yu X, Chen S, Li K. Rational design of type-I photosensitizer molecules for mitochondrion-targeted photodynamic therapy. J Mater Chem B 2024; 12:3686-3693. [PMID: 38563159 DOI: 10.1039/d4tb00099d] [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: 04/04/2024]
Abstract
Photodynamic therapy (PDT) has emerged as a promising approach for tumor treatment. However, traditional type II PDT faces limitations due to its oxygen-dependent nature. Type-I photosensitizers (PSs) exhibit superiority over conventional type-II PSs owing to their diminished oxygen dependence. Nevertheless, designing effective type-I PSs remains a significant challenge. In this work, we provide a novel strategy to tune the PDT mechanism of an excited photosensitizer through aryl substituent engineering. Using S-rhodamine as the base structure, three PSs were synthesized by incorporating phenyl, furyl, or thienyl groups at the meso position. Interestingly, furyl- or thienyl-substituted S-rhodamine are type-I-dominated PSs that produce O2˙-, while phenyl S-rhodamine results in O2˙- and 1O2 through type-I and type-II mechanisms, respectively. Experimental analyses and theoretical calculations showed that the introduction of a five-membered heterocycle at the meso position promoted intersystem crossing (ISC) and electron transfer, facilitating the production of O2˙-. Furthermore, furyl- or thienyl-substituted S-rhodamine exhibited high phototoxicity at ultralow concentrations. Thienyl-substituted S-rhodamine showed promising PDT efficacy against hypoxic solid tumors. This innovative strategy provides an alternative approach to developing new type-I PSs without the necessity for creating entirely new skeletons.
Collapse
Affiliation(s)
- Jiaxin Liang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Xiaoyun Ran
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Yanhong Liu
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Xiaoqi Yu
- Asymmetric Synthesis and Chiral Technology Key Laboratory of Sichuan Province, Department of Chemistry, Xihua University, Chengdu, Sichuan 610064, China
| | - Shanyong Chen
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Kun Li
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
89
|
Wu X, Zhou Z, Li K, Liu S. Nanomaterials-Induced Redox Imbalance: Challenged and Opportunities for Nanomaterials in Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308632. [PMID: 38380505 PMCID: PMC11040387 DOI: 10.1002/advs.202308632] [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: 11/11/2023] [Revised: 01/24/2024] [Indexed: 02/22/2024]
Abstract
Cancer cells typically display redox imbalance compared with normal cells due to increased metabolic rate, accumulated mitochondrial dysfunction, elevated cell signaling, and accelerated peroxisomal activities. This redox imbalance may regulate gene expression, alter protein stability, and modulate existing cellular programs, resulting in inefficient treatment modalities. Therapeutic strategies targeting intra- or extracellular redox states of cancer cells at varying state of progression may trigger programmed cell death if exceeded a certain threshold, enabling therapeutic selectivity and overcoming cancer resistance to radiotherapy and chemotherapy. Nanotechnology provides new opportunities for modulating redox state in cancer cells due to their excellent designability and high reactivity. Various nanomaterials are widely researched to enhance highly reactive substances (free radicals) production, disrupt the endogenous antioxidant defense systems, or both. Here, the physiological features of redox imbalance in cancer cells are described and the challenges in modulating redox state in cancer cells are illustrated. Then, nanomaterials that regulate redox imbalance are classified and elaborated upon based on their ability to target redox regulations. Finally, the future perspectives in this field are proposed. It is hoped this review provides guidance for the design of nanomaterials-based approaches involving modulating intra- or extracellular redox states for cancer therapy, especially for cancers resistant to radiotherapy or chemotherapy, etc.
Collapse
Affiliation(s)
- Xumeng Wu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin150006China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
| | - Ziqi Zhou
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
- School of Medicine and HealthHarbin Institute of TechnologyHarbin150006China
| | - Kai Li
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
- School of Medicine and HealthHarbin Institute of TechnologyHarbin150006China
| | - Shaoqin Liu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin150006China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
- School of Medicine and HealthHarbin Institute of TechnologyHarbin150006China
| |
Collapse
|
90
|
Nicolás-Morala J, Alonso-Juarranz M, Barahona A, Terrén S, Cabezas S, Falahat F, Gilaberte Y, Gonzalez S, Juarranz A, Mascaraque M. Comparative response to PDT with methyl-aminolevulinate and temoporfin in cutaneous and oral squamous cell carcinoma cells. Sci Rep 2024; 14:7025. [PMID: 38528037 DOI: 10.1038/s41598-024-57624-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/20/2024] [Indexed: 03/27/2024] Open
Abstract
Cutaneous and Head and Neck squamous cell carcinoma (CSCC, HNSCC) are among the most prevalent cancers. Both types of cancer can be treated with photodynamic therapy (PDT) by using the photosensitizer Temoporfin in HNSCC and the prodrug methyl-aminolevulinate (MAL) in CSCC. However, PDT is not always effective. Therefore, it is mandatory to correctly approach the therapy according to the characteristics of the tumour cells. For this reason, we have used cell lines of CSCC (A431 and SCC13) and HNSCC (HN5 and SCC9). The results obtained indicated that the better response to MAL-PDT was related to its localization in the plasma membrane (A431 and HN5 cells). However, with Temoporfin all cell lines showed lysosome localization, even the most sensitive ones (HN5). The expression of mesenchymal markers and migratory capacity was greater in HNSCC lines compared to CSCC, but no correlation with PDT response was observed. The translocation to the nucleus of β-catenin and GSK3β and the activation of NF-κβ is related to the poor response to PDT in the HNSCC lines. Therefore, we propose that intracellular localization of GSK3β could be a good marker of response to PDT in HNSCC. Although the molecular mechanism of response to PDT needs further elucidation, this work shows that the most MAL-resistant line of CSCC is more sensitive to Temoporfin.
Collapse
Affiliation(s)
- J Nicolás-Morala
- Department of Biology, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Experimental Dermatology and Skin Biology, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, 28034, Madrid, Spain
| | - M Alonso-Juarranz
- Oral and Maxillofacial Surgery Service, Hospital Clínico San Carlos, 28040, Madrid, Spain
- Surgery Department, Faculty of Medicine, Universidad Complutense, 28040, Madrid, Spain
| | - A Barahona
- Department of Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - S Terrén
- Department of Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - S Cabezas
- Oncology Service, Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - F Falahat
- Oral and Maxillofacial Surgery Service, Hospital Clínico San Carlos, 28040, Madrid, Spain
- Surgery Department, Faculty of Medicine, Universidad Complutense, 28040, Madrid, Spain
| | - Y Gilaberte
- Department of Dermatology, Miguel Servet University Hospital, Instituto Investigación Sanitaria (IIS), Zaragoza, Aragón, Spain
| | - S Gonzalez
- Department of Experimental Dermatology and Skin Biology, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, 28034, Madrid, Spain
- Department of Medicine and Medical Specialties, Universidad de Alcalá, Madrid, Spain
| | - A Juarranz
- Department of Biology, Universidad Autónoma de Madrid, Madrid, Spain.
- Department of Experimental Dermatology and Skin Biology, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, 28034, Madrid, Spain.
| | - M Mascaraque
- Department of Biology, Universidad Autónoma de Madrid, Madrid, Spain.
- Department of Experimental Dermatology and Skin Biology, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, 28034, Madrid, Spain.
| |
Collapse
|
91
|
Janbeih Z, Gallardo-Villagrán M, Therrien B, Diab-Assaf M, Liagre B, Benov L. Cellular Uptake and Phototoxicity Optimization of Arene Ruthenium Porphyrin Derivatives. INORGANICS 2024; 12:86. [DOI: 10.3390/inorganics12030086] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
In this study, dinuclear and tetranuclear arene ruthenium porphyrins were synthesized and assessed for their potential as photosensitizers (PSs) in photodynamic therapy (PDT) using the Colo205 colon cancer cell line as a model system. Reactive oxygen species (ROS) production, cellular uptake, impact on cell viability, and mechanisms of cell death induced by the synthesized compounds were comprehensively investigated. Our results revealed that the number of arene ruthenium units, as well as zinc (Zn) metalation of the porphyrin core, significantly influenced ROS production and increased it two-folds compared to the Zn-free analogs. The uptake of tetra-substituted Zn-porphyrins by the cancer cells increased to 2.8 nmol/106 cells compared to 0.6 nmol/106 cells of the disubstituted Zn-free and Zn-chelating porphyrins. The anticancer photo-activity of the complexes, where the percentage of metabolic activity of disubstituted Zn-porphyrins decreased to 26% when Zn was inserted, was compared to disubstituted Zn-free analogs. A further decrease in metabolic activity was observed, when the number of arene ruthenium units increased in the tetra-substituted Zn-porphyrins and tetra-substituted Zn-free compounds, reaching 4% and 14% respectively. Moreover, the percentage of apoptotic cell deaths increased to 40% when Zn was inserted into disubstituted porphyrins, compared to disubstituted Zn-free analog, and 50% when the number of arene ruthenium units increased. Overall, the tetra-substituted Zn chelating porphyrins exhibited the highest PDT efficiency, followed by the di-substituted Zn-porphyrins. These findings underscore the importance of structural design in optimizing the efficacy of arene ruthenium porphyrins as PSs for PDT, offering valuable insights for the development of targeted cancer therapeutics.
Collapse
Affiliation(s)
- Zeinab Janbeih
- Biochemistry Department, Faculty of Medicine, Kuwait University, Kuwait City 12037, Kuwait
| | - Manuel Gallardo-Villagrán
- Univ. Limoges, LABCiS, UR 22722, Faculté de Pharmacie, F-87000 Limoges, France
- Institut de Chimie, Université de Neuchâtel, Avenue de Bellevaux 51, CH-2000 Neuchâtel, Switzerland
| | - Bruno Therrien
- Institut de Chimie, Université de Neuchâtel, Avenue de Bellevaux 51, CH-2000 Neuchâtel, Switzerland
| | - Mona Diab-Assaf
- Doctoral School of Sciences and Technology, Lebanese University, Hadath, Beirut 21219, Lebanon
| | - Bertrand Liagre
- Univ. Limoges, LABCiS, UR 22722, Faculté de Pharmacie, F-87000 Limoges, France
| | - Ludmil Benov
- Biochemistry Department, Faculty of Medicine, Kuwait University, Kuwait City 12037, Kuwait
| |
Collapse
|
92
|
Ryu DS, Kim JW, Lee H, Eo SJ, Kim SH, Noh JH, Kim Y, Kang S, Na K, Park JH, Kim DH. Localized Photodynamic Therapy Using a Chlorin e6-Embedded Silicone-Covered Self-Expandable Metallic Stent as a Palliative Treatment for Malignant Esophageal Strictures. ACS Biomater Sci Eng 2024; 10:1869-1879. [PMID: 38291563 DOI: 10.1021/acsbiomaterials.3c01211] [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: 02/01/2024]
Abstract
Localized photodynamic therapy (PDT) uses a polymeric-photosensitizer (PS)-embedded, covered self-expandable metallic stent (SEMS). PDT is minimally invasive and a noteworthy potential alternative for treating esophageal strictures, where surgery is not a viable option. However, preclinical evidence is insufficient, and optimized irradiation energy dose ranges for localized PDT are unclear. Herein, we validated the irradiation energy doses of the SEMS (embedded in a PS using chlorin e6 [Ce6] and covered in silicone) and PDT-induced tissue changes in a rat esophagus. Cytotoxicity and phototoxicity in the Ce6-embedded SEMS piece with laser irradiation were significantly higher than that of the silicone-covered SEMS with or without laser and the Ce6-embedded silicone-covered SEMS without laser groups (all p < 0.001). Moreover, surface morphology, atomic changes, and homogeneous coverage of the Ce6-embedded silicone-covered membrane were confirmed. The ablation range of the porcine liver was proportionally increased with the irradiation dose (all p < 0.001). The ablation region was identified at different irradiation energy doses of 50, 100, 200, and 400 J/cm2. The in vivo study in the rat esophagus comprised a control group and 100, 200, and 400 J/cm2 energy-dose groups. Finally, histology and immunohistochemistry (TUNEL and Ki67) confirmed that the optimized Ce6-embedded silicone-covered SEMS with selected irradiation energy doses (200 and 400 J/cm2) effectively damaged the esophageal tissue without ductal perforation. The polymeric PS-embedded silicone-covered SEMS can be easily placed via a minimally invasive approach and represents a promising new approach for the palliative treatment of malignant esophageal strictures.
Collapse
Affiliation(s)
- Dae Sung Ryu
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Ji Won Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Hyeonseung Lee
- Department of Biotechnology, Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Seong Jin Eo
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Song Hee Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Jin Hee Noh
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Yuri Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Seokin Kang
- Department of Internal Medicine, Ilsan Paik Hospital, Inje University College of Medicine, 170, Juhwa-ro, Ilsanseo-gu, Goyang, Gyeonggi-do 10380, Republic of Korea
| | - Kun Na
- Department of Biotechnology, Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Jung-Hoon Park
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Do Hoon Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| |
Collapse
|
93
|
Deng W, Shang H, Tong Y, Liu X, Huang Q, He Y, Wu J, Ba X, Chen Z, Chen Y, Tang K. The application of nanoparticles-based ferroptosis, pyroptosis and autophagy in cancer immunotherapy. J Nanobiotechnology 2024; 22:97. [PMID: 38454419 PMCID: PMC10921615 DOI: 10.1186/s12951-024-02297-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 01/02/2024] [Indexed: 03/09/2024] Open
Abstract
Immune checkpoint blockers (ICBs) have been applied for cancer therapy and achieved great success in the field of cancer immunotherapy. Nevertheless, the broad application of ICBs is limited by the low response rate. To address this issue, increasing studies have found that the induction of immunogenic cell death (ICD) in tumor cells is becoming an emerging therapeutic strategy in cancer treatment, not only straightly killing tumor cells but also enhancing dying cells immunogenicity and activating antitumor immunity. ICD is a generic term representing different cell death modes containing ferroptosis, pyroptosis, autophagy and apoptosis. Traditional chemotherapeutic agents usually inhibit tumor growth based on the apoptotic ICD, but most tumor cells are resistant to the apoptosis. Thus, the induction of non-apoptotic ICD is considered to be a more efficient approach for cancer therapy. In addition, due to the ineffective localization of ICD inducers, various types of nanomaterials have been being developed to achieve targeted delivery of therapeutic agents and improved immunotherapeutic efficiency. In this review, we briefly outline molecular mechanisms of ferroptosis, pyroptosis and autophagy, as well as their reciprocal interactions with antitumor immunity, and then summarize the current progress of ICD-induced nanoparticles based on different strategies and illustrate their applications in the cancer therapy.
Collapse
Affiliation(s)
- Wen Deng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuan Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
94
|
Sayala J, Srivastava E, Kumar P, Shukla N, Kumar A, Patra AK. Photocytotoxic kinetically stable ruthenium(II)- N, N-donor polypyridyl complexes of oxalate with anticancer activity against HepG2 liver cancer cells. Dalton Trans 2024; 53:4580-4597. [PMID: 38349214 DOI: 10.1039/d3dt04058e] [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: 03/06/2024]
Abstract
Liver cancer is one of the leading causes of death that motivating scientists worldwide to synthesize novel chemotherapeutics. Ru(II)-polypyridyl complexes are extensively studied for possible therapeutic and cellular applications due to their tunable coordination chemistry, structural diversity, ligand-exchange kinetics, accessible redox states, and rich photophysical or photochemical properties. Herein, we have synthesized a series of Ru(II) polypyridyl complexes [RuII(N^N)2(ox)] (1-3), where ox is oxalate (C2O42-) and N^N is 1,10-phenanthroline (phen) (1), dipyrido[3,2-d:2',3'-f]quinoxaline (dpq) (2), and dipyrido[3,2,-a:2',3'-c]phenazine (dppz) (3). Oxalate (ox2-) was opted as a bioactive dioxo ligand to prevent facile hydrolysis in aqueous media, thereby increasing the stability of the Ru(II)-polypyridyl complexes in physiological media. We thoroughly characterized all the complexes using ESI-MS, FT-IR, UV-vis, and 1H NMR spectroscopy and other physicochemical methods. The complexes were stable under physiological conditions and under low-energy green LED light (λirr = 530 nm). However, the photoirradiation of complexes resulted in the efficient generation of singlet oxygen (1O2) as a major reactive oxygen species (ROS). The role of the extended aromatic conjugation of the N^N-donor ligands in the complexes was demonstrated by their binding propensities with CT-DNA and bovine serum albumin (BSA). Both DNA intercalation and groove binding were evidenced, while tryptophan (Trp) and tyrosine (Tyr) binding site preferences were revealed from the synchronous fluorescence spectra (SFS) of BSA. The cytotoxic profiling of the complexes performed on hepatocellular carcinoma cells (HepG2) in the dark and in the presence of green light indicated their dose-dependent cytotoxicity. The [RuII(N^N)2(ox)] complexes exhibited enhanced photocytotoxicity mediated by efficient generation of cytotoxic 1O2 and effective interaction with DNA. All the complexes were internalized by the HepG2 liver cancer cells efficiently and localized to the cytoplasm and nucleus. The complexes exhibited potent anti-proliferative, anti-clonogenic, and anti-migratory effects on the cancer cells, suggesting their potential for therapeutic applications.
Collapse
Affiliation(s)
- Juhi Sayala
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India.
| | - Ekta Srivastava
- Department of Biological Science & Bioengineering Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Priyaranjan Kumar
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India.
| | - Nitin Shukla
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India.
| | - Ashok Kumar
- Department of Biological Science & Bioengineering Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
- Center for Nanosciences, Indian Institute of Technology Kanpur, Kanpur 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ashis K Patra
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India.
| |
Collapse
|
95
|
Bahrami R, Pourhajibagher M, Gharibpour F. Antimicrobial photodynamic therapy for the management of gingivitis and white spot lesions in fixed orthodontic patients: A systematic review. Int Orthod 2024; 22:100821. [PMID: 37992475 DOI: 10.1016/j.ortho.2023.100821] [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: 09/06/2023] [Revised: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 11/24/2023]
Abstract
AIM We conducted this review to evaluate the safety and efficacy of antimicrobial photodynamic therapy (aPDT) for the management of gingivitis and white spot lesions (WSLs) in fixed orthodontic patients. METHODS The PubMed/MEDLINE, Cochrane Library, Scopus, and Google Scholar databases were searched for randomized controlled trials and clinical trials assessing the clinical effectiveness of aPDT for the management of gingivitis and WSLs in fixed orthodontic patients without time limitation. Primary outcomes were the changes in clinical parameters such as DIAGNOdent, plaque index (PI), bleeding on probing (BOP), and gingival index (GI). Secondary outcomes included measurements of microbial and inflammatory factors, such as cytokine levels (tumor necrosis factor alpha [TNF-α], interleukin-1 beta [IL-1β], and interleukin-6 [IL-6]), and bacterial counts. RESULTS Our search yielded a total of 12 studies that met the inclusion criteria. Among the 11 studies that evaluated gingivitis, the majority employed a diode laser (670nm, 150 mW, 22J/cm2, 60seconds) as the light source and methylene blue at a concentration of 0.0005% (applied for 3minutes) as the photosensitizer in a single treatment session. The included studies reported positive effects of aPDT on gingivitis management, with more improvements observed in PI, BOP, and GI following aPDT treatment. Additionally, aPDT was found to reduce the counts of periopathogens such as Porphyromonas gingivalis, as well as inflammatory factors (TNF-α, IL-1β, and IL-6). Two studies demonstrated that aPDT, particularly when administered in multiple sessions, effectively controlled the extent of WSLs during orthodontic treatment and yielded favorable outcomes that persisted for several months after treatment. CONCLUSION Based on the available evidence, aPDT appears to be a safe and effective treatment option for managing WSLs and gingivitis in patients with fixed orthodontic appliances. However, further high-quality RCTs are necessary to investigate the impact of potential confounding factors on the efficacy of aPDT.
Collapse
Affiliation(s)
- Rashin Bahrami
- Dental Sciences Research Center, Department of Orthodontics, School of Dentistry, Guilan University of Medical Sciences, Rasht, Iran.
| | - Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fateme Gharibpour
- Dental Sciences Research Center, Department of Orthodontics, School of Dentistry, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
96
|
Belykh DV, Pylina YI, Kustov AV, Startseva OM, Belykh ES, Smirnova NL, Shukhto OV, Berezin DB. Photosensitizing effects and physicochemical properties of chlorophyll a derivatives with hydrophilic oligoethylene glycol fragments at the macrocycle periphery. Photochem Photobiol Sci 2024; 23:409-420. [PMID: 38319518 DOI: 10.1007/s43630-023-00527-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/19/2023] [Indexed: 02/07/2024]
Abstract
In this work, screening studies of the cytotoxic effect of chlorins with fragments of di-, tri-, and pentaethylene glycol at the macrocycle periphery in relation to HeLa, A549, and HT29 cells were performed. It is shown that, despite different hydrophobicity, all the compounds studied have a comparable photodynamic effect. The conjugate of chlorin e6 with pentaethylene glycol, which has the lowest tendency to association among the studied compounds with tropism for low density lipoproteins and the best characteristics of the formation of molecular complexes with Tween 80, has a significant difference in dark and photoinduced toxicity (ratio IC50(dark)/IC50(photo) approximately 2 orders of magnitude for all cell lines), which allows to hope for a sufficiently large "therapeutic window". A study of the interaction of this compound with HeLa cells shows that the substance penetrates the cell and, after red light irradiation induces ROS appearance inside the cell, associated, apparently, with the photogeneration of singlet oxygen. These data indicate that photoinduced toxic effects are caused by damage to intracellular structures as a result of oxidative stress. Programmed type of cell death characterized with caspase-3 induction is prevailing. So, the conjugate of chlorin e6 with pentaethylene glycol is a promising antitumor PS that can be successfully solubilized with Tween 80, which makes it suitable for further in vivo studies.
Collapse
Affiliation(s)
- D V Belykh
- Institute of Chemistry, Komi Scientific Center, Ural Branch of the Russian Academy of Sciences, 48, Pervomaiskaya St., Syktyvkar, 167982, Russia.
| | - Y I Pylina
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the Russian Academy of Sciences, 28, Kommunisticheskaya St., Syktyvkar, 167982, Russian Federation
| | - A V Kustov
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences (ISC RAS), 1, Akademicheskaya St., 153045, Ivanovo, Russian Federation
| | - O M Startseva
- Pitirim Sorokin Syktyvkar State University, 55, Oktyabrskiy Pr., Syktyvkar, 167001, Russian Federation
| | - E S Belykh
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the Russian Academy of Sciences, 28, Kommunisticheskaya St., Syktyvkar, 167982, Russian Federation
| | - N L Smirnova
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences (ISC RAS), 1, Akademicheskaya St., 153045, Ivanovo, Russian Federation
| | - O V Shukhto
- Institute of Macroheterocyclic Compounds, Ivanovo State University of Chemistry and Technology (ISUCT), 7, Sheremetevskiy Ave., 153012, Ivanovo, Russian Federation
| | - D B Berezin
- Institute of Macroheterocyclic Compounds, Ivanovo State University of Chemistry and Technology (ISUCT), 7, Sheremetevskiy Ave., 153012, Ivanovo, Russian Federation
| |
Collapse
|
97
|
Ballestín A, Armocida D, Ribecco V, Seano G. Peritumoral brain zone in glioblastoma: biological, clinical and mechanical features. Front Immunol 2024; 15:1347877. [PMID: 38487525 PMCID: PMC10937439 DOI: 10.3389/fimmu.2024.1347877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Glioblastoma is a highly aggressive and invasive tumor that affects the central nervous system (CNS). With a five-year survival rate of only 6.9% and a median survival time of eight months, it has the lowest survival rate among CNS tumors. Its treatment consists of surgical resection, subsequent fractionated radiotherapy and concomitant and adjuvant chemotherapy with temozolomide. Despite the implementation of clinical interventions, recurrence is a common occurrence, with over 80% of cases arising at the edge of the resection cavity a few months after treatment. The high recurrence rate and location of glioblastoma indicate the need for a better understanding of the peritumor brain zone (PBZ). In this review, we first describe the main radiological, cellular, molecular and biomechanical tissue features of PBZ; and subsequently, we discuss its current clinical management, potential local therapeutic approaches and future prospects.
Collapse
Affiliation(s)
- Alberto Ballestín
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| | - Daniele Armocida
- Human Neurosciences Department, Neurosurgery Division, Sapienza University, Rome, Italy
| | - Valentino Ribecco
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| | - Giorgio Seano
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| |
Collapse
|
98
|
Liang J, Han J, Zhuang Y, Chen G, Li Y. Mitochondria-Associated Transcriptome Profiling via Localizable Aggregation-Induced Emission Photosensitizers in Live Cells. ACS Chem Biol 2024; 19:419-427. [PMID: 38264802 DOI: 10.1021/acschembio.3c00617] [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: 01/25/2024]
Abstract
In recent decades, there has been increasing interest in studying mitochondria through transcriptomic research. Various exogenous fusion protein-based proximity labeling methods have been reported that focus on the site of one particular protein/peptide and might also influence the corresponding localization or interactome. To enable unbiased and high spatial-resolution profiling of mitochondria-associated transcriptomes in live cells, a flexible RNA proximity labeling approach was developed using aggregation-induced emission (AIE) type photosensitizers (PSs) that possess great mitochondria-targeting capabilities. Their accumulation in an enclosed mitochondrial environment tends to enhance the fluorescence emission and reactive oxygen species generation. By comparing the in vitro optical properties, photosensitization processes, as well as the in cellulo mitochondrial specificity and RNA labeling performance of four AIE PSs, high-throughput sequencing analysis was conducted using TFPy-mediated RNA proximity labeling in live HeLa cells. This approach successfully captured a comprehensive list of transcripts, including mitochondria-encoded RNAs, as well as some nuclear-derived RNAs located at the outer mitochondrial membrane and interacting organelles. This small molecule-based proximity labeling method bypasses complex genetic manipulation and transfection steps, making it readily applicable for diverse research purposes.
Collapse
Affiliation(s)
- Jiying Liang
- Department of Chemistry, The University of Hong Kong, Hong Kong 999077, China
| | - Jinghua Han
- Department of Chemistry, The University of Hong Kong, Hong Kong 999077, China
| | - Yuan Zhuang
- Department of Chemistry, The University of Hong Kong, Hong Kong 999077, China
- Hong Kong Quantum AI Lab Limited, Hong Kong 999077, China
| | - GuanHua Chen
- Department of Chemistry, The University of Hong Kong, Hong Kong 999077, China
- Hong Kong Quantum AI Lab Limited, Hong Kong 999077, China
| | - Ying Li
- Department of Chemistry, The University of Hong Kong, Hong Kong 999077, China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, New Territories, Hong Kong 999077, China
| |
Collapse
|
99
|
Ghosh C, Ali LMA, Bessin Y, Clément S, Richeter S, Bettache N, Ulrich S. Self-assembled porphyrin-peptide cages for photodynamic therapy. Org Biomol Chem 2024; 22:1484-1494. [PMID: 38289387 DOI: 10.1039/d3ob01887c] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
The development of photodynamic therapy requires access to smart photosensitizers which combine appropriate photophysical and biological properties. Interestingly, supramolecular and dynamic covalent chemistries have recently shown their ability to produce novel architectures and responsive systems through simple self-assembly approaches. Herein, we report the straightforward formation of porphyrin-peptide conjugates and cage compounds which feature on their surface chemical groups promoting cell uptake and specific organelle targeting. We show that they self-assemble, in aqueous media, into positively-charged nanoparticles which generate singlet oxygen upon green light irradiation, while also undergoing a chemically-controlled disassembly due to the presence of reversible covalent linkages. Finally, the biological evaluation in cells revealed that they act as effective photosensitizers and promote synergistic effects in combination with Doxorubicin.
Collapse
Affiliation(s)
- Chandramouli Ghosh
- Institut des Biomolécules Max Mousseron (IBMM), Université of Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Lamiaa M A Ali
- Institut des Biomolécules Max Mousseron (IBMM), Université of Montpellier, CNRS, ENSCM, Montpellier, France.
- Department of Biochemistry Medical Research Institute, University of Alexandria, 21561 Alexandria, Egypt
| | - Yannick Bessin
- Institut des Biomolécules Max Mousseron (IBMM), Université of Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Sébastien Clément
- Institut Charles Gerhardt Montpellier (ICGM), Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | - Sébastien Richeter
- Institut Charles Gerhardt Montpellier (ICGM), Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | - Nadir Bettache
- Institut des Biomolécules Max Mousseron (IBMM), Université of Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Sébastien Ulrich
- Institut des Biomolécules Max Mousseron (IBMM), Université of Montpellier, CNRS, ENSCM, Montpellier, France.
| |
Collapse
|
100
|
Gonzalo-Navarro C, Zafon E, Organero JA, Jalón FA, Lima JC, Espino G, Rodríguez AM, Santos L, Moro AJ, Barrabés S, Castro J, Camacho-Aguayo J, Massaguer A, Manzano BR, Durá G. Ir(III) Half-Sandwich Photosensitizers with a π-Expansive Ligand for Efficient Anticancer Photodynamic Therapy. J Med Chem 2024; 67:1783-1811. [PMID: 38291666 PMCID: PMC10859961 DOI: 10.1021/acs.jmedchem.3c01276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/12/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024]
Abstract
One approach to reduce the side effects of chemotherapy in cancer treatment is photodynamic therapy (PDT), which allows spatiotemporal control of the cytotoxicity. We have used the strategy of coordinating π-expansive ligands to increase the excited state lifetimes of Ir(III) half-sandwich complexes in order to facilitate the generation of 1O2. We have obtained derivatives of formulas [Cp*Ir(C∧N)Cl] and [Cp*Ir(C∧N)L]BF4 with different degrees of π-expansion in the C∧N ligands. Complexes with the more π-expansive ligand are very effective photosensitizers with phototoxic indexes PI > 2000. Furthermore, PI values of 63 were achieved with red light. Time-dependent density functional theory (TD-DFT) calculations nicely explain the effect of the π-expansion. The complexes produce reactive oxygen species (ROS) at the cellular level, causing mitochondrial membrane depolarization, cleavage of DNA, nicotinamide adenine dinucleotide (NADH) oxidation, as well as lysosomal damage. Consequently, cell death by apoptosis and secondary necrosis is activated. Thus, we describe the first class of half-sandwich iridium cyclometalated complexes active in PDT.
Collapse
Affiliation(s)
- Carlos Gonzalo-Navarro
- Departamento
de Química Inorgánica, Orgánica y Bioquímica-
IRICA, Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Avda. C. J. Cela, 10, 13071 Ciudad Real, Spain
| | - Elisenda Zafon
- Departament
de Biologia, Facultat de Ciències, Universitat de Girona, Maria Aurèlia Capmany 40, 17003 Girona, Spain
| | - Juan Angel Organero
- Departamento
de Química Física, Facultad de Ciencias Ambientales
y Bioquímicas and INAMOL, Universidad
de Castilla-La Mancha, 45071 Toledo, Spain
| | - Félix A. Jalón
- Departamento
de Química Inorgánica, Orgánica y Bioquímica-
IRICA, Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Avda. C. J. Cela, 10, 13071 Ciudad Real, Spain
| | - Joao Carlos Lima
- LAQV-REQUIMTE,
Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Gustavo Espino
- Departamento
de Química, Facultad de Ciencias, Universidad de Burgos, Pza. Misael Bañuelos, s/n, 09001 Burgos, Spain
| | - Ana María Rodríguez
- Departamento
de Química Inorgánica, Orgánica y Bioquímica-
IRICA, Escuela Técnica Superior de Ingenieros Industriales, Universidad de Castilla-La Mancha, Avda. C. J. Cela, 3, 13071 Ciudad Real, Spain
| | - Lucía Santos
- Departamento
de Química Física, Facultad de Ciencias y Tecnologías
Químicas, Universidad de Castilla-La
Mancha, Avda. C. J. Cela,
s/n, 13071 Ciudad
Real, Spain
| | - Artur J. Moro
- LAQV-REQUIMTE,
Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Sílvia Barrabés
- Departament
de Biologia, Facultat de Ciències, Universitat de Girona, Maria Aurèlia Capmany 40, 17003 Girona, Spain
| | - Jessica Castro
- Departament
de Biologia, Facultat de Ciències, Universitat de Girona, Maria Aurèlia Capmany 40, 17003 Girona, Spain
| | - Javier Camacho-Aguayo
- Analytical
Chemistry Department, Analytic Biosensors Group, Instituto de Nanociencia
y Nanomateriales de Aragon, Faculty of Sciences, University of Zaragoza, 50009 Zaragoza, Spain
| | - Anna Massaguer
- Departament
de Biologia, Facultat de Ciències, Universitat de Girona, Maria Aurèlia Capmany 40, 17003 Girona, Spain
| | - Blanca R. Manzano
- Departamento
de Química Inorgánica, Orgánica y Bioquímica-
IRICA, Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Avda. C. J. Cela, 10, 13071 Ciudad Real, Spain
| | - Gema Durá
- Departamento
de Química Inorgánica, Orgánica y Bioquímica-
IRICA, Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Avda. C. J. Cela, 10, 13071 Ciudad Real, Spain
| |
Collapse
|