Next Article in Journal
Consideration of SHP-1 as a Molecular Target for Tumor Therapy
Previous Article in Journal
Studying the Roles of the Renin–Angiotensin System in Accelerating the Disease of High-Fat-Diet-Induced Diabetic Nephropathy in a db/db and ACE2 Double-Gene-Knockout Mouse Model
Previous Article in Special Issue
Perfluorocarbon Nanoemulsions with Fluorous Chlorin-Type Photosensitizers for Antitumor Photodynamic Therapy in Hypoxia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Editorial: Photodynamic Therapy as an Important Tool for Biological Breakthroughs—Photoactive Photosensitizers Applied from Cancer to Microbial Targets

by
Leandro M. O. Lourenço
1,*,
Augusto C. Tomé
1 and
João P. C. Tomé
2
1
LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
2
CQE, IMS, DEQ, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(1), 330; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25010330
Submission received: 3 December 2023 / Accepted: 25 December 2023 / Published: 26 December 2023
(This article belongs to the Special Issue Photodynamic Therapy as Important Tool for Biological Breakthroughs)

1. Introduction

Photodynamic therapy (PDT) stands as an approved clinical treatment for both oncologic and nononcologic disorders. It boasts various advantages when compared to traditional oncological approaches like chemotherapy, radiotherapy, and surgery [1,2]. The generation of cytotoxic reactive oxygen species (ROS), especially singlet oxygen (1O2), through the interaction of harmless photoactivable molecules, known as photosensitizers (PS), with oxygen, brings about the demise of cancer cells while maintaining a favorable cosmetic outcome [3,4]. Notwithstanding the notable advancements achieved through PS-mediated PDT, this therapeutic approach encounters certain constraints. These include issues such as PS aggregation, limited solubility in physiological fluids, and specificity toward tissues or cells [5]. The development of innovative PS that may overcome these limitations is, thus, an important target.
Besides this, it is important to highlight that antimicrobial photodynamic therapy (aPDT) has been also explored as an innovative therapeutic approach because it can be used to inactivate a variety of microbial forms (e.g., bacteria [6,7,8], fungi [9,10,11], and viruses [12,13,14]) without causing significant damage to host tissues and without the development of resistance to the photosensitization process [15].
Lately, numerous research endeavors centered around novel molecules have been undertaken to enhance the selectivity and outcome of PDT using versatile PS [16]. These dyes exhibit the capacity to overcome the constraints associated with conventional PS. They not only enhance the solubility in biological environments but also bolster the PS uptake in target areas [17]. This indicates a substantial potential for advancements in the next iterations of PS agents [18], heralding the advent of a promising new generation of PS.
The subject of investigation, titled ‘Photodynamic Therapy as Important Tool for Biological Breakthroughs’, encompasses a collection of ten manuscripts that delve into the latest advancements within the field of PDT, focusing on its applications against cancer, bacteria, and viral targets.
Markova and co-workers (contribution 1) explored perfluorocarbon nanoemulsions with fluorous chlorin-type PS for antitumor PDT in hypoxia conditions (Table 1, entry 1). The authors developed three versions of oxygen-rich chlorin molecules and used them to create nanoemulsions containing perfluorodecalin. These formulations included hydrophobic PS for absorbing specific light wavelengths and an oxygen carrier. These changes did not affect chlorin’s photosensitizing properties, including the generation of cytotoxic ROS. The emulsions entered colon carcinoma cells (HCT116) and accumulated in mitochondria. When these cells loaded with emulsions were illuminated, lipids underwent rapid peroxidation, and plasma membranes were compromised, resulting in cell death (photonecrosis). Significantly, in PDT conditions, the emulsions effectively sensitized cells cultured under prolonged hypoxia and those with acute hypoxia induced by oxygen depletion. Emulsions caused notably more photodamage in hypoxic conditions compared to situations without emulsions. With minimal dark cytotoxicity, these materials show promise as efficient and biocompatible tools for PDT-enhanced elimination of hypoxic cells.
Abrahamse and co-workers (contribution 2) emphasized the cytotoxic effects of combinative PDT effect using zinc(II) tetrasulfonic phthalocyanine (ZnPcS4) and cannabidiol (CBD) on a cervical cancer cell line (HeLa cells)—Table 1, entry 2. The authors emphasized the anticancer properties of CBD, an isolate from the Cannabis sativa L. plant. They found that CBD can inhibit tumor growth and its spread. To assess these effects, they conducted experiments using HeLa CC-cultured cells in vitro. These cells were exposed to varying doses of ZnPcS4 in combination with CBD. When HeLa CC cells received the lowest predetermined half-maximal inhibitory concentration dose (ICD50) of 0.125 µM ZnPcS4 along with 0.5 µM CBD and were subjected to laser irradiation, they exhibited a highly significant and advantageous form of cell death. However, further investigation into this promising treatment strategy is required.
Abrahamse and co-workers (contribution 3) also researched the synthesis, characterization, and PDT efficacy of the aluminum(III) chloride tetramercaptoacetate phthalocyanine (AlClPcTS41). They investigated the impact of AlClPcTS41 alone and when combined with PEGylated copper-gold bimetallic nanoparticles (PEG-CuAuNPs) as PS on colon cancer cells (Caco-2)—Table 1, entry 3. To create AlClPcTS41-PEG-CuAuNPs, they covalently linked AlClPcTS41 to PEG-CuAuNPs using an amide bond. Subsequently, they examined the subcellular uptake, cellular proliferation, and the antitumor PDT effects of AlClPcTS41, PEG-CuAuNPs, and AlClPcTS41-PEG-CuAuNPs in vitro using Caco-2 cells. Both the designed AlClPcTS41 and AlClPcTS41-PEG-CuAuNPs exhibited significant abilities to generate ROS, resulting in a substantial reduction in cell viability after PDT treatment.
Shigekawa and co-workers (contribution 4) evidenced a cell-level analysis to visualize the PDT process using porphylipoprotein and talaporphyrin sodium (Table 1, entry 4). Their research uncovered the contrasting mechanisms of PDT between two PS: porphylipoprotein (PLP), which has garnered recent attention for its potential in cancer treatment, and talaporphyrin sodium (NPe6). NPe6 was found to accumulate in lysosomes, whereas PLP was incorporated into phagosomes formed through PLP injection. In PDT, NPe6 generated ROS, leading to the production of actin filaments and stress fibers. However, with PLP, ROS generated by PDT remained confined within the phagosomes until the phagosomal membrane was disrupted. This delay influenced the initiation of Ras homolog family member A (RhoA) activation and RhoA*/Rho-associated coiled-coil containing kinases (ROCK) generation. Once the phagosomal membrane was disrupted, ROS were released, accelerating the production of actin filaments, stress fibers, and causing blebbing earlier than observed with NPe6. PLP also increased the elastic modulus of cells without RhoA activity in the early stage because phagosomes played a role in polymerizing actin filaments and forming pseudopodia. Given PLP’s high selectivity and uptake by cancer cells, a more substantial PDT effect can be anticipated by skillfully combining these discovered characteristics, particularly the rapid onset of a strong effect in the early stages.
Terreno and co-workers (contribution 5) have developed a prostate-specific membrane antigen (PSMA)-targeted probe designed for near-infrared fluorescence (NIRF)-guided surgery and PDT, with synthesis and preclinical validation as their primary focus. The researchers conjugated the PSMA-617 binding motif to a silicon(IV) phthalocyanine (designated by IRDye700DX-NHS), and this conjugation did not alter the photophysical characteristics of the fluorophore (Table 1, entry 5). The affinity of IRDye700DX-PSMA-617 toward prostate cancer (PCa) cells correlated with their PSMA expression levels. NIRF imaging demonstrated significant tumor uptake after injecting 1 or 5 nmol, resulting in a high tumor-to-muscle ratio. Flow cytometry and confocal images confirmed the co-localization of PSMA+ cells with IRDye700DX-PSMA uptake. Notably, ex vivo analysis of a tumor specimen revealed significant PSMA expression by tumor-associated macrophages, likely attributable to extracellular vesicles secreted by PSMA+ tumor cells. In PDT experiments, cell viability decreased in a concentration-dependent manner (from 75% at 10 nM to 12% at 500 nM), while the controls exhibited no cytotoxicity. This research has yielded a PSMA-targeted NIRF dye with dual imaging-PDT capabilities.
Malinowski and co-workers (contribution 6) embarked on a study exploring a method for producing glycosylated porphyrins through nucleophilic aromatic substitution (SNAr), coupled with investigations into cellular uptake (Table 1, entry 6). Glycoporphyrins constitute a valuable group of compounds, particularly significant for applications in PDT and various other biomedical uses. Despite substantial advancements in this field, there is a growing need for innovative and diverse synthetic approaches to create carbohydrate–porphyrin hybrids. In this work, the authors introduced an efficient, gentle, and metal-free method for crafting glycoporphyrins. The versatility of the SNAr procedure is demonstrated through 16 distinct examples. Additionally, preliminary biological assessments have been conducted to evaluate the cytotoxicity and cellular uptake of these molecules.
Hu and co-workers (contribution 7) have presented a study showcasing the precise eradication of malignant tumor cells through sinoporphyrin sodium-mediated PDT (Table 1, entry 7). In this research, human malignant glioblastoma U-118MG and melanoma A375 cells were chosen as targets. The product of the power density, irradiation time, and PS concentration was denoted as the total photodynamic parameter. This parameter was employed to investigate the mechanisms of PS sinoporphyrin sodium (DVDMS)-mediated PDT (DVDMS-PDT). The findings indicated a negative correlation between the survival rates of U-118MG and A375 cells and the total photodynamic parameter value in the curve. Remarkably, this correlation conformed to an exponential function. Furthermore, by manipulating the three parameters at random, the authors achieved controllable tumor cell-killing effects, thus validating the accuracy and reproducibility of their formula. The establishment and application of a functional relationship among PDT parameters play a pivotal role in predicting PDT outcomes and offering precise treatment.
Lourenço and co-workers (contribution 8) reported a study that evaluated the antifungal and sporicidal photodynamic activity of two tetra- and octa-β-substituted zinc(II) phthalocyanine dyes with dimethylaminopyridinium groups at the periphery and their quaternized derivatives (Table 1, entry 8). All phthalocyanine derivatives were prepared and assessed as PS for their effects on Fusarium oxysporum conidia. Antimicrobial photoinactivation experiments were performed with each PS at 0.1, 1, 10, and 20 µM under white light irradiation at an irradiance of 135 mW·cm–2, for 60 min (light dose of 486 J·cm−2). High aPDT efficiency was observed for the prepared PS (10 µM). The quaternized PS showed better aPDT performance than the non-quaternized ones, even at the low concentration of 1 µM, and a light dose of 486 J·cm−2. These cationic phthalocyanines are potent photodynamic PS for antifungal applications due to their ability to effectively inactivate resistant forms, like conidia, with low concentrations and reasonable energy doses.
Lourenço and co-workers (contribution 9) investigated the in vitro photoinactivation of Fusarium oxysporum conidia using light-activated ammonium phthalocyanines (Table 1, entry 9). This research examined the photodynamic antifungal and sporicidal capabilities of tetra- and octa-substituted phthalocyanines containing ammonium groups. The experiments involved conducting aPDT tests with varying PS concentrations while exposing them to white light. Both PS demonstrated high aPDT efficiency, reaching the detection limit. Among the two PS, the tetra-substituted variant proved to be the most effective, requiring the lowest concentration and the shortest irradiation time to completely inactivate the conidia (40 µM, 30 min, 243 J·cm−2). While the octa-substituted PS also achieved complete inactivation, it required a longer irradiation period and a higher concentration (60 µM, 60 min, 486 J·cm−2). These findings are significant because they indicate that these phthalocyanines exhibit potent antifungal photodynamic properties, demanding low concentrations and moderate energy doses to effectively combat resilient biological forms.
Lourenço and co-workers (contribution 10) documented their research on the PDT for combating T4-like bacteriophages, serving as a model for mammalian viruses in blood. This study investigated the antiviral effects of three cationic porphyrins (Table 1, entry 10) when subjected to white light exposure, and the outcomes were compared to the performance of the approved methylene blue (MB). Under isotonic conditions, none of the PS induced hemolysis when employing the T4-like phage as a surrogate for mammalian viruses. In the context of plasma, all porphyrins exhibited superior efficacy in aPDT compared to MB for inactivating the T4-like phage. Furthermore, the most efficient PS demonstrated a moderate inactivation rate for the T4-like phage in whole blood. Nevertheless, these porphyrins, much like MB, show promise as safe PS for photoinactivating viruses in blood plasma.
The Editors anticipate that the contributions gathered in this Research Topic will have a meaningful influence on the field by providing fresh insights into the research, development, and understanding of the use of remarkable dyes in the PDT approach.

Author Contributions

L.M.O.L. was responsible for the analysis, interpretation of the results, and contributed to the manuscript preparation. A.C.T. was responsible for the analysis, interpretation of the results, and contributed to the manuscript preparation. J.P.C.T. was responsible for the analysis, interpretation of the results, and contributed to the manuscript preparation. L.M.O.L., A.C.T. and J.P.C.T. were also responsible for funding acquisition and resources. All authors have read and agreed to the published version of the manuscript.

Funding

This work received financial support from PT national funds (FCT/MCTES, Fundação para a Ciência e a Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior) through projects LAQV-REQUIMTE (UIDB/50006/2020 and UIDP/50006/2020), CQE (UIDB/00100/2020 + UIDP/00100/2020), IMS (LA/P/0056/2020) and P2020-PTDC/QUI-QOR/31770/2017.

Acknowledgments

The authors thank the University of Aveiro and FCT/MCTES (Fundação para a Ciência e a Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior) for the financial support to LAQV-REQUIMTE (UIDB/50006/2020 and UIDP/50006/2020), CQE (UIDB/00100/2020 and UIDP/00100/2020), and IMS (LA/P/0056/2020) funded by FCT/MCTES through national funds. The authors also acknowledge FCT/MCTES for the national funds received through project P2020-PTDC/QUI-QOR/31770/2017.

Conflicts of Interest

The authors declare no conflicts of interest.

List of Contributions

  • Nguyen, M.T.; Guseva, E.V.; Ataeva, A.N.; Sigan, A.L.; Shibaeva, A.V.; Dmitrieva, M.V.; Burtsev, I.D.; Volodina, Y.L.; Radchenko, A.S.; Egorov, A.E.; et al. Perfluorocarbon Nanoemulsions with Fluorous Chlorin-Type Photosensitizers for Antitumor Photodynamic Therapy in Hypoxia. Int. J. Mol. Sci. 2023, 24, 7995. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24097995.
  • Razlog, R.; Kruger, C.A.; Abrahamse, H. Cytotoxic Effects of Combinative ZnPcS4 Photosensitizer Photodynamic Therapy (PDT) and Cannabidiol (CBD) on a Cervical Cancer Cell Line. Int. J. Mol. Sci. 2023, 24, 6151. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24076151.
  • Simelane, N.W.N.; Matlou, G.G.; Abrahamse, H. Photodynamic Therapy of Aluminum Phthalocyanine Tetra Sodium 2-Mercaptoacetate Linked to PEGylated Copper–Gold Bimetallic Nanoparticles on Colon Cancer Cells. Int. J. Mol. Sci. 2023, 24, 1902. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24031902.
  • Kamiyanagi, M.; Taninaka, A.; Ugajin, S.; Nagoshi, Y.; Kurokawa, H.; Ochiai, T.; Arashida, Y.; Takeuchi, O.; Matsui, H.; Shigekawa, H. Cell-Level Analysis Visualizing Photodynamic Therapy with Porphylipoprotein and Talaporphyrin Sodium. Int. J. Mol. Sci. 2022, 23, 13140. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232113140.
  • Capozza, M.; Stefania, R.; Dinatale, V.; Bitonto, V.; Conti, L.; Grange, C.; Skovronova, R.; Terreno, E. A Novel PSMA-Targeted Probe for NIRF-Guided Surgery and Photodynamic Therapy: Synthesis and Preclinical Validation. Int. J. Mol. Sci. 2022, 23, 12878. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232112878.
  • Rosa, M.; Jędryka, N.; Skorupska, S.; Grabowska-Jadach, I.; Malinowski, M. New Route to Glycosylated Porphyrins via Aromatic Nucleophilic Substitution (SNAr)—Synthesis and Cellular Uptake Studies. Int. J. Mol. Sci. 2022, 23, 11321. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231911321.
  • Lv, G.; Dong, Z.; Zhao, Y.; Ma, N.; Jiang, X.; Li, J.; Wang, J.; Wang, J.; Zhang, W.; Lin, X.; et al. Precision Killing of Sinoporphyrin Sodium-Mediated Photodynamic Therapy against Malignant Tumor Cells. Int. J. Mol. Sci. 2022, 23, 10561. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231810561.
  • Lourenço, L.M.O.; Cunha, Â.; Sierra-Garcia, I.N. Light-Driven Tetra- and Octa-β-substituted Cationic Zinc(II) Phthalocyanines for Eradicating Fusarium oxysporum Conidia. Int. J. Mol. Sci. 2023, 24, 16980. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms242316980.
  • Gamelas, S.R.D.; Sierra-Garcia, I.N.; Tomé, A.C.; Cunha, Â.; Lourenço, L.M.O. In Vitro Photoinactivation of Fusarium Oxysporum Conidia with Light-Activated Ammonium Phthalocyanines. Int. J. Mol. Sci. 2023, 24, 3922. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24043922.
  • Santos, P.; Gomes, A.T.P.C.; Lourenço, L.M.O.; Faustino, M.A.F.; Neves, M.G.P.M.S.; Almeida, A. Anti-Viral Photodynamic Inactivation of T4-like Bacteriophage as a Mammalian Virus Model in Blood. Int. J. Mol. Sci. 2022, 23, 11548. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231911548.

References

  1. Pham, T.C.; Nguyen, V.-N.; Choi, Y.; Lee, S.; Yoon, J. Recent Strategies to Develop Innovative Photosensitizers for Enhanced Photodynamic Therapy. Chem. Rev. 2021, 121, 13454–13619. [Google Scholar] [CrossRef] [PubMed]
  2. Olszowy, M.; Nowak-Perlak, M.; Woźniak, M. Current Strategies in Photodynamic Therapy (PDT) and Photodynamic Diagnostics (PDD) and the Future Potential of Nanotechnology in Cancer Treatment. Pharmaceutics 2023, 15, 1712. [Google Scholar] [CrossRef] [PubMed]
  3. Halaskova, M.; Rahali, A.; Almeida-Marrero, V.; Machacek, M.; Kucera, R.; Jamoussi, B.; Torres, T.; Novakova, V.; de la Escosura, A.; Zimcik, P. Peripherally Crowded Cationic Phthalocyanines as Efficient Photosensitizers for Photodynamic Therapy. ACS Med. Chem. Lett. 2021, 12, 502–507. [Google Scholar] [CrossRef] [PubMed]
  4. Du, Y.; Guo, Y.; Xiao, X.; Zhu, X.; Liu, Y.; Ruan, X.; Huang, X.; Yuan, Y.; Wei, X. Glutathione Depletion and Photosensitizer Activation Augments Efficacy of Tumor Photodynamic Immunotherapy. Chem. Eng. J. 2022, 442, 136170. [Google Scholar] [CrossRef]
  5. Gunaydin, G.; Gedik, M.E.; Ayan, S. Photodynamic Therapy for the Treatment and Diagnosis of Cancer—A Review of the Current Clinical Status. Front. Chem. 2021, 9, 686303. [Google Scholar] [CrossRef] [PubMed]
  6. Drescher, K.; Nadell, C.D.; Stone, H.A.; Wingreen, N.S.; Bassler, B.L. Solutions to the Public Goods Dilemma in Bacterial Biofilms. Curr. Biol. 2014, 24, 50–55. [Google Scholar] [CrossRef] [PubMed]
  7. Bottery, M.J.; Pitchford, J.W.; Friman, V.-P. Ecology and Evolution of Antimicrobial Resistance in Bacterial Communities. ISME J. 2021, 15, 939–948. [Google Scholar] [CrossRef] [PubMed]
  8. Kim, G.; Karbaschi, M.; Cooke, M.; Gaitas, A. Light-Based Methods for Whole Blood Bacterial Inactivation Enabled by a Recirculating Flow System. Photochem. Photobiol. 2018, 94, 744–751. [Google Scholar] [CrossRef] [PubMed]
  9. Sierra-Garcia, I.N.; Cunha, Â.; Lourenço, L.M.O. In Vitro Photodynamic Treatment of Fusarium oxysporum Conidia through the Action of Thiopyridinium and Methoxypyridinium Chlorins. J. Photochem. Photobiol. A Chem. 2022, 432, 114081. [Google Scholar] [CrossRef]
  10. Ziental, D.; Mlynarczyk, D.T.; Czarczynska-Goslinska, B.; Lewandowski, K.; Sobotta, L. Photosensitizers Mediated Photodynamic Inactivation against Fungi. Nanomaterials 2021, 11, 2883. [Google Scholar] [CrossRef] [PubMed]
  11. Yaakoub, H.; Mina, S.; Calenda, A.; Bouchara, J.-P.; Papon, N. Oxidative Stress Response Pathways in Fungi. Cell. Mol. Life Sci. 2022, 79, 333. [Google Scholar] [CrossRef] [PubMed]
  12. Costa, L.; Faustino, M.A.F.; Neves, M.G.P.M.S.; Cunha, Â.; Almeida, A. Photodynamic Inactivation of Mammalian Viruses and Bacteriophages. Viruses 2012, 4, 1034–1074. [Google Scholar] [CrossRef] [PubMed]
  13. Ke, M.-R.; Eastel, J.M.; Ngai, K.L.K.; Cheung, Y.-Y.; Chan, P.K.S.; Hui, M.; Ng, D.K.P.; Lo, P.-C. Photodynamic Inactivation of Bacteria and Viruses Using Two Monosubstituted Zinc(II) Phthalocyanines. Eur. J. Med. Chem. 2014, 84, 278–283. [Google Scholar] [CrossRef] [PubMed]
  14. Sharshov, K.; Solomatina, M.; Kurskaya, O.; Kovalenko, I.; Kholina, E.; Fedorov, V.; Meerovich, G.; Rubin, A.; Strakhovskaya, M. The Photosensitizer Octakis(Cholinyl)Zinc Phthalocyanine with Ability to Bind to a Model Spike Protein Leads to a Loss of SARS-CoV-2 Infectivity In Vitro When Exposed to Far-Red LED. Viruses 2021, 13, 643. [Google Scholar] [CrossRef] [PubMed]
  15. Ribeiro, C.P.S.; Lourenço, L.M.O. Overview of Cationic Phthalocyanines for Effective Photoinactivation of Pathogenic Microorganisms. J. Photochem. Photobiol. C Photochem. Rev. 2021, 48, 100422. [Google Scholar] [CrossRef]
  16. Lu, Y.; Sun, W.; Du, J.; Fan, J.; Peng, X. Immuno-Photodynamic Therapy (IPDT): Organic Photosensitizers and Their Application in Cancer Ablation. JACS Au. 2023, 3, 682–699. [Google Scholar] [CrossRef] [PubMed]
  17. Rodrigues, J.A.; Correia, J.H. Enhanced Photodynamic Therapy: A Review of Combined Energy Sources. Cells 2022, 11, 3995. [Google Scholar] [CrossRef] [PubMed]
  18. Moura, N.M.M.; Monteiro, C.J.P.; Tomé, A.C.; Neves, M.G.P.M.S.; Cavaleiro, J.A.S. Synthesis of Chlorins and Bacteriochlorins from Cycloaddition Reactions with Porphyrins. Arkivoc 2022, 2022, 54–98. [Google Scholar] [CrossRef]
Table 1. Photoactive photosensitizers applied from cancer to microbial targets.
Table 1. Photoactive photosensitizers applied from cancer to microbial targets.
EntryPhotosensitizer StructuresApplication
1Ijms 25 00330 i001PDT in colon carcinoma cells (HCT116)
2Ijms 25 00330 i002PDT in cervical cancer cell line (HeLa cells)
3Ijms 25 00330 i003PDT in colon cancer cells (Caco-2)
4Ijms 25 00330 i004Studies in lysosomes or phagosomes
5Ijms 25 00330 i005PDT in prostate cancer (PCa) cells
6Ijms 25 00330 i006Studies in A549 and MCF-7 cellular uptake
7Ijms 25 00330 i007PDT in human malignant glioblastoma U-118MG and melanoma A375 cells
8Ijms 25 00330 i008aPDT in Fusarium
oxysporum conidia
9Ijms 25 00330 i009aPDT in Fusarium
oxysporum conidia
10Ijms 25 00330 i010aPDT in T4-like
bacteriophages
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lourenço, L.M.O.; Tomé, A.C.; Tomé, J.P.C. Editorial: Photodynamic Therapy as an Important Tool for Biological Breakthroughs—Photoactive Photosensitizers Applied from Cancer to Microbial Targets. Int. J. Mol. Sci. 2024, 25, 330. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25010330

AMA Style

Lourenço LMO, Tomé AC, Tomé JPC. Editorial: Photodynamic Therapy as an Important Tool for Biological Breakthroughs—Photoactive Photosensitizers Applied from Cancer to Microbial Targets. International Journal of Molecular Sciences. 2024; 25(1):330. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25010330

Chicago/Turabian Style

Lourenço, Leandro M. O., Augusto C. Tomé, and João P. C. Tomé. 2024. "Editorial: Photodynamic Therapy as an Important Tool for Biological Breakthroughs—Photoactive Photosensitizers Applied from Cancer to Microbial Targets" International Journal of Molecular Sciences 25, no. 1: 330. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25010330

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop