Fighting Cancer with Photodynamic Therapy and Nanotechnologies: Current Challenges and Future Directions
Abstract
1. Introduction
1.1. Biological Characteristics of Neoplasms
1.2. Current Cancer Statistics and Future Projections
1.3. Challenges in Drug Development and Potential Solutions
1.4. Role of Artificial Intelligence (AI) and Quantum Computing in Oncology
1.5. Evolution of Cancer Therapies
1.6. Innovations in Photodynamic Therapy (PDT)
1.7. Future Directions in Cancer Research
2. State-of-the-Art Nano-Photosensitizers, Functional Nanosystems and Nanoscale Delivery Vehicles
2.1. Addressing Oxygen Deficiency in Tumor Environments
2.2. Two-Photon Excited Fluorescence for Deeper Tumor Imaging
2.3. Covalent Organic Frameworks (COFs) for Photodynamic Therapy
2.4. Multifunctional Nanoplatforms for Targeted Tumor Management
2.5. Carbon Dots for Photodynamic Therapy
2.6. Oxygen-Generating PDT Nanoagents
2.7. Lanthanide-Triplet Near-Infrared (NIR) Sensitization
2.8. Oxygen-Independent PDT via RNA-Targeting Photosensitizers
2.9. BODIPY-Based Photosensitizers for PDT
2.10. Advances in PDT for Glioma and Deep Tumors
3. Nano-Photosensitizers in PDT Targeting Mitochondria in Cancer Cells
3.1. Mechanisms of PDT and Challenges in Solid Tumors
3.2. Advances in Nano-Photosensitizers for Hypoxia Mitigation
3.3. Novel Photosensitizer Strategies to Improve PDT
3.4. Mitochondrial Targeting for Enhanced PDT in Breast Cancer
3.5. Future Perspectives on Mitochondrial Modulation in PDT
3.6. Mitochondrial Complex III and ROS Release
3.7. Atovaquone and Its Impact on Mitochondrial Respiration
3.8. Biomimetic Nanoparticles in Cancer Treatment
3.9. Photosensitizers and Aggregation-Induced Emission (AIE)
3.10. Design of Organic AIEgens for Cancer Therapy
3.11. Mitochondrial Targeting in Cancer Therapy
3.12. Cytotoxicity of PDT and Targeting Mitochondria
3.13. DNA Nanotechnology for Mitochondrial-Targeted PDT
3.14. Mitochondria-Targeted Photosensitizers in Cancer Therapy
3.15. Colorectal Cancer and PDT
3.16. Challenges in PDT Efficacy
3.17. Overcoming Biological Barriers in PDT
3.18. Immune Response in PDT
3.19. PROTAC Nanoplatforms for PDT
3.20. Induction of Immunogenic Cell Death (ICD)
4. Final Remarks and Conclusions
4.1. Recent Advancements in PDT: New Approaches
4.2. Future Directions and Innovations in PDT
4.3. The Role of Mitochondria in Cancer Cells and PDT
4.4. Mechanisms of ROS Production and Its Impact on Cellular Health
4.5. PDT with Mitochondrial Targeting: A Promising Approach for Cancer Treatment
4.6. Challenges and Advances in PDT for Solid Tumors
4.7. Future Directions: Mitochondrial-Targeted Nano-Photosensitizers
4.8. Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
Ace-DSB | acetal-terminated distyrylbenzene derivative |
ACQ | aggregation-induced quenching |
AI | artificial intelligence |
AIE | aggregation-induced emission |
AIEgens | aggregation-induced emission luminogens |
ALA | 5-aminolevulinic acid |
Ald-DSB | aldehyde-terminated molecules |
AS-AMD | poly-AS1411 aptamer |
ATO | atovaquone |
ATP | adenosine triphosphate |
BDF | bonding defects functionalization |
BET | extra-terminal domain |
BGM | buffalo green monkey kidney |
BODIPY | boron-dipyrromethene |
BODIPY-CDs complexes | BODIPY-α-CD, BODIPY-β-CD, and BODIPY-γ-CD where CD = cyclodextrin |
3BP | 3-bromopyruvate |
BPN | black phosphorus nanosheets |
BPN/MnO2 | nanosponge from tightly packed MnO2-laden BPN |
BPN/MnO2/DOX | BPN/MnO2 nanocomposite loaded with DOX |
BRD4 | bromodomain-containing protein 4 |
CAT@MnPFs | catalase-loaded manganese-porphyrin frameworks |
CD | cyclodextrin |
CDs | carbon dots |
Ce6 | chlorin e6 |
CIII | mitochondrial complex III; cytochrome bc1 complex; ubiquinol cytochrome c reductase; |
CMPNs | self-assembly of hyaluronic acid-conjugated-methoxy poly(ethylene glycol)-diethylenetriamine-grafted-(chlorin e6-dihydrolipoic acid-(3-carboxypropyl)triphenylphosphine bromide) polymeric ligands (HA-c-mPEG-Deta-g-(Ce6-DHLA-TPP)) and NaErF4:Tm@NaYF4 core-shell UCNPs |
COFs | covalent organic frameworks |
COUPY 1 | coumarin-based fluorophores 1 |
COUPY 2 | coumarin-based fluorophores 2 |
CPPO | bis [2,4,5-trichloro-6-(pentyloxycar-bonyl)phenyl]oxalate |
C-TBD NPs | C-TBD nanoparticles |
CTPP | triphenylphosphine |
dBET6 | small-molecule degrader |
DCs | dendritic cells |
DEM | diethyl maleate |
DNA | deoxyribonucleic acid |
DOX | doxorubicin |
EHMONs | eccentric hollow mesoporous organic silica NPs |
EMT | epithelial-mesenchymal transition |
ENBS-B | molecular superoxide radical (O2–•) generator |
ETC | electro-transport chain |
FDA | U.S. Food and Drug Administration |
FEITC | β-phenylethyl isothiocyanate |
FFRKGPLGLAGC-PEG-DSPE | DSPE-PEG2000-modified transformable peptides |
FLI | fluorescence imaging |
FRET | fluorescence resonance energy transfer |
FR/NIR | far-red/near-infrared |
GSDME | gasdermin E |
GSH | glutathione |
H2O2 | hydrogen peroxide |
HeLa | cervical cancer cell line containing human papillomavirus 18 (HPV-18) |
HIF-1-alpha | hypoxia-inducible factor 1-alpha |
HPV-18 | human papillomavirus 18 |
ICB | immune checkpoint blockade |
ICD | immunogenic cell death |
ICG | indocyanine green |
Ir | iridium |
IR780 | IR-780 dye for near-infrared fluorescence imaging/photosensitizer |
MB | methylene blue |
MC | Mn3 [Co(CN) 6]2 |
MC-r | Mn3 [Co(CN) 6]2 -Ru |
MCF-7 | human breast cancer cell line |
MDA-MB-231 | TSPO-positive breast cancer cells |
mETC | mitochondrial electron transport chain |
MKN-45 cells | human gastric adenocarcinoma cell line |
MRI | magnetic resonance imaging |
mROS | mitochondrial ROS |
MSNs | mesoporous silica nanoparticles |
Mn3O4@MSNs@IR780 | NPs-PS (IR780)—90 nm MSNs—capping surface pores 5 nm Mn3O4 NPs |
mtDAMPs | mitochondrial damage-associated molecular patterns |
mtDNA | mitochondrial DNA |
MTCS | HeLa multicellular tumor spheroids |
NaGdF4:Nd | nanocrystals |
NBEX (X = S, Se, Te) | type-III PS designed to bind to RNA |
NCs | nanocapsules |
NCOFs | boron-dipyrromethene (BODIPY)-decorated nanoscale COFs |
NIR | near infrared |
nMOL | nanoscale metal-organic layer |
NO | nitric oxide |
NP | nanoparticle |
NPs | nanoparticles |
NSs | nanoshuttles |
1O2 | singlet oxygen |
OTL38 | pafolacianine |
OxgeMCC-r SAE | OxgeMCC-r single-atom enzyme (SAE) |
OXPHOS | oxidative phosphorylation |
2PA | two-photon absorption |
PD-1 | programmed cell death-1 |
PD-L1 | programmed death ligand 1 |
PDT | photodynamic therapy |
PFC | perfluorocarbon |
PFCs | perfluorocarbons |
PLGA | polylactic-co-glycolic acid |
PpIX | Protoporphyrin IX |
PROTAC | proteolysis-targeting chimeras |
PTT | photothermal therapy |
RBCm | red blood cell membrane |
RBS | Roussin’s black salt |
Rh-PTZ NPs | Rh-PTZ nanoparticles |
ROS | reactive oxygen species |
SAE | single-atom enzyme |
SBU | secondary building units |
SRRF | super-resolution radial fluctuations |
TAMs | tumor-associated M2 macrophages |
TBD | TPE-BT-DC |
TCA | tricarboxylic acid cycle |
TIME | tumor immune microenvironment |
TME | tumor microenvironment |
TNBC | triple-negative breast cancer |
TPP-TK-PPa | amphiphilic heterodimeric photosensitizer |
TSPO | translocator protein |
UCNPs | upconversion nanoparticles |
UCNPs@mSiO2-NH2&RBS | platform with nitric oxide release upon NIR irradiation |
US | United States |
VEGF | vascular endothelial growth factor |
WHO | World Health Organization |
ZnPc | zinc phthalocyanine |
ΔΨm | mitochondrial membrane potential |
Increased | ↑ |
Decreased | ↓ |
Present | + |
Absent/Missing | - |
References
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
- American Cancer Society. Cancer Facts & Figures 2024. Basic Cancer Facts. Available online: https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2024/2024-cancer-facts-and-figures-acs.pdf (accessed on 27 December 2024).
- World Health Organization. Eastern Mediterranean Region. Available online: https://www.emro.who.int/media/news/world-cancer-day-2024.html (accessed on 27 December 2024).
- Prasad, V.; Mailankody, S. Research and Development Spending to Bring a Single Cancer Drug to Market and Revenues After Approval. JAMA Intern. Med. 2017, 177, 1569–1575. [Google Scholar] [CrossRef] [PubMed]
- Mohs, R.C.; Greig, N.H. Drug discovery and development: Role of basic biological research. Alzheimer’s Dement. 2017, 3, 651–657. [Google Scholar] [CrossRef]
- Sun, D.; Gao, W.; Hu, H.; Zhou, S. Why 90% of clinical drug development fails and how to improve it? Acta Pharm. Sinica. B 2022, 12, 3049–3062. [Google Scholar] [CrossRef]
- Wang, F.; Ruan, D.Y.; Xu, R.H. Challenges and opportunities in oncology drug development and clinical research in China. Cell 2024, 187, 1578–1583. [Google Scholar] [CrossRef]
- Pei, Z. Computer-aided drug discovery: From traditional simulation methods to language models and quantum computing. Cell Rep. Phys. Sci. 2024, 5, 102334. [Google Scholar] [CrossRef]
- Huanbutta, K.; Burapapadh, K.; Kraisit, P.; Sriamornsak, P.; Ganokratanaa, T.; Suwanpitak, K.; Sangnim, T. Artificial intelligence-driven pharmaceutical industry: A paradigm shift in drug discovery, formulation development, manufacturing, quality control, and post-market surveillance. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2024, 203, 106938. [Google Scholar] [CrossRef]
- Nissan, N.; Allen, M.C.; Sabatino, D.; Biggar, K.K. Future Perspective: Harnessing the Power of Artificial Intelligence in the Generation of New Peptide Drugs. Biomolecules 2024, 14, 1303. [Google Scholar] [CrossRef]
- Swanton, C.; Bernard, E.; Abbosh, C.; André, F.; Auwerx, J.; Balmain, A.; Bar-Sagi, D.; Bernards, R.; Bullman, S.; DeGregori, J.; et al. Embracing cancer complexity: Hallmarks of systemic disease. Cell 2024, 187, 1589–1616. [Google Scholar] [CrossRef]
- Falzone, L.; Salomone, S.; Libra, M. Evolution of Cancer Pharmacological Treatments at the Turn of the Third Millennium. Front. Pharmacol. 2018, 9, 1300. [Google Scholar] [CrossRef]
- McAleer, S. A history of cancer and its treatment: Presidential Address to the Ulster Medical Society. 7th October 2021. Ulst. Med. J. 2022, 91, 124–129. [Google Scholar]
- Sonkin, D.; Thomas, A.; Teicher, B.A. Cancer treatments: Past, present, and future. Cancer Genet. 2024, 286, 18–24. [Google Scholar] [CrossRef] [PubMed]
- Ailioaie, L.M.; Ailioaie, C.; Litscher, G. Synergistic Nanomedicine: Photodynamic, Photothermal and Photoimmune Therapy in Hepatocellular Carcinoma: Fulfilling the Myth of Prometheus? Int. J. Mol. Sci. 2023, 24, 8308. [Google Scholar] [CrossRef]
- Kessel, D. Photodynamic Therapy: A Brief History. J. Clin. Med. 2019, 8, 1581. [Google Scholar] [CrossRef] [PubMed]
- Gunaydin, G.; Gedik, M.E.; Ayan, S. Photodynamic Therapy-Current Limitations and Novel Approaches. Front. Chem. 2021, 9, 691697. [Google Scholar] [CrossRef]
- Grin, M.; Suvorov, N.; Ostroverkhov, P.; Pogorilyy, V.; Kirin, N.; Popov, A.; Sazonova, A.; Filonenko, E. Advantages of Combined Photodynamic Therapy in the Treatment of Oncological Diseases. Biophys. Rev. 2022, 14, 941–963. [Google Scholar] [CrossRef]
- Huis in ‘t Veld, R.V.; Heuts, J.; Ma, S.; Cruz, L.J.; Ossendorp, F.A.; Jager, M.J. Current Challenges and Opportunities of Photodynamic Therapy against Cancer. Pharmaceutics 2023, 15, 330. [Google Scholar] [CrossRef]
- Mariño-Ocampo, N.; Dibona-Villanueva, L.; Escobar-Álvarez, E.; Guerra-Díaz, D.; Zúñiga-Núñez, D.; Fuentealba, D.; Robinson-Duggon, J. Recent Photosensitizer Developments, Delivery Strategies and Combination-based Approaches for Photodynamic Therapy. Photochem. Photobiol. 2023, 2, 469–497. [Google Scholar] [CrossRef]
- Ailioaie, L.M.; Ailioaie, C.; Litscher, G. Latest Innovations and Nanotechnologies with Curcumin as a Nature-Inspired Photosensitizer Applied in the Photodynamic Therapy of Cancer. Pharmaceutics 2021, 13, 1562. [Google Scholar] [CrossRef]
- Ailioaie, L.M.; Litscher, G. Curcumin and Photobiomodulation in Chronic Viral Hepatitis and Hepatocellular Carcinoma. Int. J. Mol. Sci. 2020, 21, 7150. [Google Scholar] [CrossRef]
- Wang, X.; Peng, J.; Meng, C.; Feng, F. Recent advances for enhanced photodynamic therapy: From new mechanisms to innovative strategies. Chem. Sci. 2024, 15, 12234–12257. [Google Scholar] [CrossRef]
- Mao, D.; Wu, W.; Ji, S.; Chen, C.; Hu, F.; Kong, D.; Ding, D.; Liu, B. Chemiluminescence-Guided Cancer Therapy Using a Chemiexcited Photosensitizer. Chem 2017, 3, 991–1007. [Google Scholar] [CrossRef]
- Li, M.; Xia, J.; Tian, R.; Wang, J.; Fan, J.; Du, J.; Long, S.; Song, X.; Foley, J.W.; Peng, X. Near-Infrared Light-Initiated Molecular Superoxide Radical Generator: Rejuvenating Photodynamic Therapy against Hypoxic Tumors. J. Am. Chem. Soc. 2018, 140, 14851–14859. [Google Scholar] [CrossRef]
- Sun, C.L.; Li, J.; Wang, X.Z.; Shen, R.; Liu, S.; Jiang, J.Q.; Li, T.; Song, Q.W.; Liao, Q.; Fu, H.B.; et al. Rational Design of Organic Probes for Turn-On Two-Photon Excited Fluorescence Imaging and Photodynamic Therapy. Chem 2019, 5, 600–616. [Google Scholar] [CrossRef]
- Guan, Q.; Fu, D.D.; Li, Y.A.; Kong, X.M.; Wei, Z.Y.; Li, W.Y.; Zhang, S.J.; Dong, Y.B. BODIPY-Decorated Nanoscale Covalent Organic Frameworks for Photodynamic Therapy. iScience 2019, 14, 180–198. [Google Scholar] [CrossRef]
- Wu, Q.; Chen, G.; Gong, K.; Wang, J.; Ge, X.; Liu, X.; Guo, S.; Wang, F. MnO2-Laden Black Phosphorus for MRI-Guided Synergistic PDT, PTT, and Chemotherapy. Matter 2019, 1, 496–512. [Google Scholar] [CrossRef]
- Wei, S.M.; Feng, K.; Li, C.; Xie, N.; Wang, Y.; Yang, X.L.; Chen, B.; Tung, C.H.; Wu, L.Z. ZnCl2 Enabled Synthesis of Highly Crystalline and Emissive Carbon Dots with Exceptional Capability to Generate O2.–. Matter 2020, 2, 495–506. [Google Scholar] [CrossRef]
- Wang, D.; Wu, H.; Phua, S.Z.F.; Yang, G.; Qi Lim, W.; Gu, L.; Qian, C.; Wang, H.; Guo, Z.; Chen, H.; et al. Self-assembled single-atom nanozyme for enhanced photodynamic therapy treatment of tumor. Nat. Commun. 2020, 11, 357. [Google Scholar] [CrossRef]
- Zheng, B.; Zhong, D.; Xie, T.; Zhou, J.; Li, W.; Ilyas, A.; Lu, Y.; Zhou, M.; Deng, R. Near-infrared photosensitization via direct triplet energy transfer from lanthanide nanoparticles. Chem 2021, 7, 1615–1625. [Google Scholar] [CrossRef]
- Yao, Q.; Fan, J.; Long, S.; Zhao, X.; Li, H.; Du, J.; Shao, K.; Peng, X. The concept and examples of type-III photosensitizers for cancer photodynamic therapy. Chem 2022, 8, 197–209. [Google Scholar] [CrossRef]
- Gao, D.; Zhang, B.; Liu, Y.; Hu, D.; Sheng, Z.; Zhang, X.; Yuan, Z. Molecular Engineering of Near-Infrared Light-Responsive BODIPY-Based Nanoparticles with Enhanced Photothermal and Photoacoustic Efficiencies for Cancer Theranostics. Theranostics 2019, 9, 5315–5331. [Google Scholar] [CrossRef]
- Dartar, S.; Ucuncu, M.; Karakus, E.; Hou, Y.; Zhao, J.; Emrullahoglu, M. BODIPY-Vinyl Dibromides as Triplet Sensitisers for Photodynamic Therapy and Triplet-Triplet Annihilation Upconversion. Chem. Commun. 2021, 57, 6039–6042. [Google Scholar] [CrossRef]
- Wang, J.; Jiang, Z.; Huang, C.; Zhao, S.; Zhu, S.; Liu, R.; Zhu, H. Self-Assembled BODIPY Nanoparticles for Near-Infrared Fluorescence Bioimaging. Molecules 2023, 28, 2997. [Google Scholar] [CrossRef]
- Das, S.; Dey, S.; Patra, S.; Bera, A.; Ghosh, T.; Prasad, B.; Sayala, K.D.; Maji, K.; Bedi, A.; Debnath, S. BODIPY-Based Molecules for Biomedical Applications. Biomolecules 2023, 13, 1723. [Google Scholar] [CrossRef]
- Lu, B.; Lu, X.; Mu, M.; Meng, S.; Feng, Y.; Zhang, Y. Novel near-infrared BODIPY-cyclodextrin complexes for photodynamic therapy. Heliyon 2024, 10, e26907. [Google Scholar] [CrossRef]
- Lee, J.H.; Wee, C.W. Treatment of Adult Gliomas: A Current Update. Brain Neurorehabil. 2022, 15, e24. [Google Scholar] [CrossRef]
- Yang, K.; Wu, Z.; Zhang, H.; Zhang, N.; Wu, W.; Wang, Z.; Dai, Z.; Zhang, X.; Zhang, L.; Peng, Y.; et al. Glioma targeted therapy: Insight into future of molecular approaches. Mol. Cancer. 2022, 21, 39. [Google Scholar] [CrossRef]
- Simon, M.; Hagemann, A.; Gajadin, S.; Signorelli, F.; Vincent, A.J.P.E. Surgical treatment for insular gliomas. A systematic review and meta-analysis on behalf of the EANS neuro-oncology section. Brain Spine 2024, 4, 102828. [Google Scholar] [CrossRef]
- Lucke-Wold, B.; Rangwala, B.S.; Shafique, M.A.; Siddiq, M.A.; Mustafa, M.S.; Danish, F.; Nasrullah, R.M.U.; Zainab, N.; Haseeb, A. Focus on current and emerging treatment options for glioma: A comprehensive review. World J. Clin. Oncol. 2024, 15, 482–495. [Google Scholar] [CrossRef]
- Kim, M.M.; Darafsheh, A. Light Sources and Dosimetry Techniques for Photodynamic Therapy. Photochem. Photobiol. 2020, 96, 280–294. [Google Scholar] [CrossRef]
- Xiang, M.; Zhou, Q.; Shi, Z.; Wang, X.; Li, M.; Jia, Y.; Li, S.; Yang, F.; Wang, W.; Chen, T.; et al. A Review of Light Sources and Enhanced Targeting for Photodynamic Therapy. Curr. Med. Chem. 2021, 28, 6437–6457. [Google Scholar] [CrossRef]
- Li, L.; Yang, J.H.; Fa, X.M.; Liu, M.S.; Wang, Q.L.; Zeng, T.F.; Chen, R.Z.; Ou, J.; Xia, X.W. Preliminary investigation of nitric oxide release from upconverted nanoparticles excited at 808 nm near-infrared for brain tumors. Heliyon 2024, 10, e33576. [Google Scholar] [CrossRef]
- Alimoradi, H.; Greish, K.; Gamble, A.B.; Giles, G.I. Controlled Delivery of Nitric Oxide for Cancer Therapy. Pharm. Nanotechnol. 2019, 7, 279–303. [Google Scholar] [CrossRef]
- Yu, H.; Tiemuer, A.; Yao, X.; Zuo, M.; Wang, H.Y.; Liu, Y.; Chen, X. Mitochondria-specific near-infrared photoactivation of peroxynitrite upconversion luminescent nanogenerator for precision cancer gas therapy. Acta Pharm. Sin. B 2024, 14, 378–391. [Google Scholar] [CrossRef]
- Tang, Y.; Li, Q.; Zhou, Z.; Bai, H.; Xiao, N.; Xie, J.; Li, C. Nitric oxide-based multi-synergistic nanomedicine: An emerging therapeutic for anticancer. J. Nanobiotechnol. 2024, 22, 674. [Google Scholar] [CrossRef]
- Qamar, M.; Basharat, A.; Qamar, S.A.; Bilal, M.; Franco, M.; Iqbal, H. Enzyme-loaded nanostructured materials for the degradation of environmental pollutants. Curr. Opin. Environ. Sci. Health 2022, 30, 100400. [Google Scholar] [CrossRef]
- Du, B.; Tung, C.H. Enzyme-Assisted Photodynamic Therapy Based on Nanomaterials. ACS Biomater. Sci. Eng. 2020, 6, 2506–2517. [Google Scholar] [CrossRef]
- Qiao, Y.; Tang, X.; Qiuju, X.; Zhang, G. Enzyme-loaded manganese-porphyrin metal-organic nanoframeworks for oxygen-evolving photodynamic therapy of hypoxic cells. Heliyon 2024, 10, e33902. [Google Scholar] [CrossRef]
- Darvin, P.; Chandrasekharan, A.; Varadarajan, S.N.; Chandrasekhar, L.; Maliakkal, R.T.; SM, J.S.; Varghese Jancy, S.; Santhoshkumar, T.R. Mitochondria targeted redox GFP reveals time and dose dependent onset and progression of mitochondrial oxidation with diverging cell death decisions during photodynamic therapy. Photodiagnosis Photodyn. Ther. 2020, 31, 101921. [Google Scholar] [CrossRef]
- Yang, Z.; Wang, J.; Ai, S.; Sun, J.; Mai, X.; Guan, W. Self-generating oxygen enhanced mitochondrion-targeted photodynamic therapy for tumor treatment with hypoxia scavenging. Theranostics 2019, 9, 6809–6823. [Google Scholar] [CrossRef]
- Wen, J.; Luo, Y.; Gao, H.; Zhang, L.; Wang, X.; Huang, J.; Shang, T.; Zhou, D.; Wang, D.; Wang, Z.; et al. Mitochondria-targeted nanoplatforms for enhanced photodynamic therapy against hypoxia tumor. J. Nanobiotechnol. 2021, 19, 440. [Google Scholar] [CrossRef]
- Nash, G.T.; Luo, T.; Lan, G.; Ni, K.; Kaufmann, M.; Lin, W. Nanoscale Metal-Organic Layer Isolates Phthalocyanines for Efficient Mitochondria-Targeted Photodynamic Therapy. J. Am. Chem. Soc. 2021, 143, 2194–2199. [Google Scholar] [CrossRef]
- Cai, X.; Wang, K.N.; Ma, W.; Yang, Y.; Chen, G.; Fu, H.; Cui, C.; Yu, Z.; Wang, X. Multifunctional AIE iridium (III) photosensitizer nanoparticles for two-photon-activated imaging and mitochondria targeting photodynamic therapy. J. Nanobiotechnol. 2021, 19, 254. [Google Scholar] [CrossRef]
- Vagia, E.; Mahalingam, D.; Cristofanilli, M. The Landscape of Targeted Therapies in TNBC. Cancers 2020, 12, 916. [Google Scholar] [CrossRef]
- Sun, X.; Wang, M.; Wang, M.; Yu, X.; Guo, J.; Sun, T.; Li, X.; Yao, L.; Dong, H.; Xu, Y. Metabolic Reprogramming in Triple-Negative Breast Cancer. Front. Oncol. 2020, 10, 428. [Google Scholar] [CrossRef]
- World Health Organization. Breast Cancer, 13 March 2024. Available online: https://www.who.int/news-room/fact-sheets/detail/breast-cancer (accessed on 15 January 2025).
- Huang, Z.; Li, D.; Gou, F.; Xian, T.; Hu, S.H.; Xu, J.; Luo, F.Y.; Chen, Z.Z.; Wang, C.B.; Zang, M.Y. Mitochondria-targeted photosensitizer based nanoplatform loading glutathione inhibitor for enhanced breast cancer photodynamic therapy. Colloids Surf. B Biointerfaces 2022, 220, 112956. [Google Scholar] [CrossRef]
- Seah, D.; Cheng, Z.; Vendrell, M. Fluorescent Probes for Imaging in Humans: Where Are We Now? ACS Nano 2023, 17, 19478–19490. [Google Scholar] [CrossRef]
- Cytalux: Package Insert/Prescribing Info. Drugs.com. Last Updated on 17 October 2024. Available online: https://www.drugs.com/pro/cytalux.html (accessed on 18 January 2025).
- Ge, Y.; O’Shea, D.F. Review of Clinically Assessed Molecular Fluorophores for Intraoperative Image Guided Surgery. Molecules 2024, 29, 5964. [Google Scholar] [CrossRef]
- Bonelli, J.; Ortega-Forte, E.; Rovira, A.; Bosch, M.; Torres, O.; Cuscó, C.; Rocas, J.; Ruiz, J.; Marchán, V. Improving Photodynamic Therapy Anticancer Activity of a Mitochondria-Targeted Coumarin Photosensitizer Using a Polyurethane-Polyurea Hybrid Nanocarrier. Biomacromolecules 2022, 23, 2900–2913. [Google Scholar] [CrossRef]
- Wang, R.; Li, X.; Yoon, J. Organelle-Targeted Photosensitizers for Precision Photodynamic Therapy. ACS Appl. Mater. Interfaces 2021, 13, 19543–19571. [Google Scholar] [CrossRef]
- Chen, Q.; Vazquez, E.J.; Moghaddas, S.; Hoppel, C.L.; Lesnefsky, E.J. Production of reactive oxygen species by mitochondria: Central role of complex III. J. Biol. Chem. 2003, 278, 36027–36031. [Google Scholar] [CrossRef]
- Ma, X.; Jin, M.; Cai, Y.; Xia, H.; Long, K.; Liu, J.; Yu, Q.; Yuan, J. Mitochondrial electron transport chain complex III is required for antimycin A to inhibit autophagy. Chem. Biol. 2011, 18, 1474–1481. [Google Scholar] [CrossRef]
- Antimycin A from Streptomyces sp. Available online: https://www.sigmaaldrich.com/RO/en/product/sigma/a8674?srsltid=AfmBOoomBUZa_KNALz9iXKTqRrwX7X0lOYTphd0y_7SWH41O6E9GHb5a (accessed on 21 January 2025).
- Coates, J.T.T.; Rodriguez-Berriguete, G.; Puliyadi, R.; Ashton, T.; Prevo, R.; Wing, A.; Granata, G.; Pirovano, G.; McKenna, G.W.; Higgins, G.S. The anti-malarial drug atovaquone potentiates platinum-mediated cancer cell death by increasing oxidative stress. Cell Death Discov. 2020, 6, 110. [Google Scholar] [CrossRef]
- Ashton, T.M.; Fokas, E.; Kunz-Schughart, L.A.; Folkes, L.K.; Anbalagan, S.; Huether, M.; Kelly, C.J.; Pirovano, G.; Buffa, F.M.; Hammond, E.M.; et al. The anti-malarial atovaquone increases radiosensitivity by alleviating tumour hypoxia. Nat. Commun. 2016, 7, 12308. [Google Scholar] [CrossRef]
- Fry, M.; Pudney, M. Site of action of the antimalarial hydroxynaphthoquinone, 2-[trans-4-(4′-chlorophenyl) cyclohexyl]-3-hydroxy-1,4-naphthoquinone (566C80). Biochem. Pharmacol. 1992, 43, 1545–1553. [Google Scholar] [CrossRef]
- Mather, M.W.; Darrouzet, E.; Valkova-Valchanova, M.; Cooley, J.W.; McIntosh, M.T.; Daldal, F.; Vaidya, A.B. Uncovering the molecular mode of action of the antimalarial drug atovaquone using a bacterial system. J. Biol. Chem. 2005, 280, 27458–27465. [Google Scholar] [CrossRef]
- Nixon, G.L.; Moss, D.M.; Shone, A.E.; Lalloo, D.G.; Fisher, N.; O’Neill, P.M.; Ward, S.A.; Biagini, G.A. Antimalarial pharmacology and therapeutics of atovaquone. J. Antimicrob. Chemother. 2013, 68, 977–985. [Google Scholar] [CrossRef]
- Chen, A.; Yu, Z.; Ma, N.; Lu, X.; Zhang, Y.; Xu, W.; Wang, Y.; Xie, J.; Qin, Y.; Mo, G.; et al. Atovaquone enhances antitumor efficacy of TCR-T therapy by augmentation of ROS-induced ferroptosis in hepatocellular carcinoma. Cancer Immunol. Immunother. 2024, 73, 49. [Google Scholar] [CrossRef]
- Li, X.; Wang, H.; Li, Z.; Li, D.; Lu, X.; Ai, S.; Dong, Y.; Liu, S.; Wu, J.; Guan, W. Oxygen tank for synergistic hypoxia relief to enhance mitochondria-targeted photodynamic therapy. Biomater. Res. 2022, 26, 47. [Google Scholar] [CrossRef]
- Zeng, F.; Fan, Z.; Li, S.; Li, L.; Sun, T.; Qiu, Y.; Nie, L.; Huang, G. Tumor Microenvironment Activated Photoacoustic-Fluorescence Bimodal Nanoprobe for Precise Chemo-immunotherapy and Immune Response Tracing of Glioblastoma. ACS Nano 2023, 17, 19753–19766. [Google Scholar] [CrossRef]
- Luo, H.; Gao, S. Recent advances in fluorescence imaging-guided photothermal therapy and photodynamic therapy for cancer: From near-infrared-I to near-infrared-II. J. Control Release. 2023, 362, 425–445. [Google Scholar] [CrossRef]
- Qi, J.; Hu, X.; Dong, X.; Lu, Y.; Lu, H.; Zhao, W.; Wu, W. Towards more accurate bioimaging of drug nanocarriers: Turning aggregation-caused quenching into a useful tool. Adv. Drug Deliv. Rev. 2019, 143, 206–225. [Google Scholar] [CrossRef]
- Yanhong Duo, Y.; Xiang, Z.; Gao, G.; Luo, G.; Tang, Z.B. Biomedical application of aggregation-induced emission luminogen-based fluorescent sensors. TrAC Trends Anal. Chem. 2023, 167, 117252. [Google Scholar] [CrossRef]
- Zhao, X.; Wu, X.; Shang, R.; Chen, H.; Tan, N. A structure-guided strategy to design Golgi apparatus-targeted type-I/II aggregation-induced emission photosensitizers for efficient photodynamic therapy. Acta Biomater. 2024, 183, 235–251. [Google Scholar] [CrossRef]
- Zielonka, J.; Joseph, J.; Sikora, A.; Hardy, M.; Ouari, O.; Vasquez-Vivar, J.; Cheng, G.; Lopez, M.; Kalyanaraman, B. Mitochondria-Targeted Triphenylphosphonium-Based Compounds: Syntheses, Mechanisms of Action, and Therapeutic and Diagnostic Applications. Chem. Rev. 2017, 117, 10043–10120. [Google Scholar] [CrossRef]
- Son, S.; Kim, J.; Kim, J.; Kim, B.; Lee, J.; Kim, Y.; Li, M.; Kang, H.; Kim, J.S. Cancer therapeutics based on diverse energy sources. Chem. Soc. Rev. 2022, 51, 8201–8215. [Google Scholar] [CrossRef]
- Ganji, C.; Muppala, V.; Khan, M.; Nagaraju, P.G.; Farran, B. Mitochondrial-targeted nanoparticles: Delivery and therapeutic agents in cancer. Drug Discov. Today 2023, 28, 103469. [Google Scholar] [CrossRef]
- Long, X.; Liu, M.; Nan, Y.; Chen, Q.; Xiao, Z.; Xiang, Y.; Ying, X.; Sun, J.; Huang, Q.; Ai, K. Revitalizing Ancient Mitochondria with Nano-Strategies: Mitochondria-Remedying Nanodrugs Concentrate on Disease Control. Adv. Mater. 2024, 3, e2308239. [Google Scholar] [CrossRef]
- Zhao, H.; Li, L.; Li, F.; Liu, C.; Huang, M.; Li, J.; Gao, F.; Ruan, X.; Yang, D. An Energy-Storing DNA-Based Nanocomplex for Laser-Free Photodynamic Therapy. Adv. Mater. 2022, 34, e2109920. [Google Scholar] [CrossRef]
- Zhao, H.; Wang, Z.; Yang, S.; Zhang, R.; Guo, J.; Yang, D. Energy-storing DNA-based hydrogel remodels tumor microenvironments for laser-free photodynamic immunotherapy. Biomaterials 2024, 309, 122620. [Google Scholar] [CrossRef]
- Chen, M.; Xu, H.; Chang, P.; Li, X.; Liu, S.; Xu, L.; Xu, K.; Cheng, G. An intelligent poly aptamer-encoded DNA nanoclew for tumor site activated mitochondria-targeted photodynamic therapy and MR imaging. Mater. Today Bio. 2024, 29, 101318. [Google Scholar] [CrossRef]
- Zhang, S.; Yang, L.; Ling, X.; Shao, P.; Wang, X.; Edwards, W.B.; Bai, M. Tumor mitochondria-targeted photodynamic therapy with a translocator protein (TSPO)-specific photosensitizer. Acta Biomater. 2015, 28, 160–170. [Google Scholar] [CrossRef]
- Betlazar, C.; Middleton, R.J.; Banati, R.; Liu, G.J. The Translocator Protein (TSPO) in Mitochondrial Bioenergetics and Immune Processes. Cells 2020, 9, 512. [Google Scholar] [CrossRef]
- Li, Y.; Chen, L.; Papadopoulos, V. The mitochondrial translocator protein (TSPO, 18 kDa): A key multifunctional molecule in liver diseases. Biochimie 2024, 224, 91–103. [Google Scholar] [CrossRef]
- WHO. Colorectal Cancer. 11 July 2023. Available online: https://www.who.int/news-room/fact-sheets/detail/colorectal-cancer (accessed on 27 January 2025).
- Wang, L.; Geng, H.; Liu, Y.; Liu, L.; Chen, Y.; Wu, F.; Liu, Z.; Ling, S.; Wang, Y.; Zhou, L. Hot and cold tumors: Immunological features and the therapeutic strategies. MedComm 2023, 4, e343. [Google Scholar] [CrossRef]
- Zhou, Y.; Zhang, W.; Wang, B.; Wang, P.; Li, D.; Cao, T.; Zhang, D.; Han, H.; Bai, M.; Wang, X.; et al. Mitochondria-targeted photodynamic therapy triggers GSDME-mediated pyroptosis and sensitizes anti-PD-1 therapy in colorectal cancer. J. Immunother. Cancer 2024, 12, e008054. [Google Scholar] [CrossRef]
- Shi, L.; Hu, F.; Duan, Y.; Wu, W.; Dong, J.; Meng, X.; Zhu, X.; Liu, B. Hybrid Nanospheres to Overcome Hypoxia and Intrinsic Oxidative Resistance for Enhanced Photodynamic Therapy. ACS Nano 2020, 14, 2183–2190. [Google Scholar] [CrossRef]
- Hu, T.; Wang, Z.; Shen, W.; Liang, R.; Yan, D.; Wei, M. Recent advances in innovative strategies for enhanced cancer photodynamic therapy. Theranostics 2021, 11, 3278–3300. [Google Scholar] [CrossRef]
- Zhuang, Y.; Liu, K.; He, Q.; Gu, X.; Jiang, C.; Wu, J. Hypoxia signaling in cancer: Implications for therapeutic interventions. MedComm 2023, 4, e203. [Google Scholar] [CrossRef]
- Li, J.; Wang, Y.; Tao, J.; Su, X.; Zhu, F.; Lu, W.; Han, X.; Dang, M.; Weng, L. Mitochondria-Targeting and Oxygen Self-Supplying Eccentric Hollow Nanoplatform for Enhanced Breast Cancer Photodynamic Therapy. Therapy. Bioinorg. Chem. Appl. 2024, 2024, 6618388. [Google Scholar] [CrossRef]
- Liu, Z.H.; Mo, X.W.; Jiang, W.; Liu, C.; Yin, Y.; Yang, H.Y.; Fu, Y. Multifunctional hyaluronic acid ligand-assisted construction of CD44- and mitochondria-targeted self-assembled upconversion nanoparticles for enhanced photodynamic therapy. Dalton Trans. 2024, 53, 16885–16895. [Google Scholar] [CrossRef]
- Yuan, Z.; Fan, G.; Wu, H.; Liu, C.; Zhan, Y.; Qiu, Y.; Shou, C.; Gao, F.; Zhang, J.; Yin, P.; et al. Photodynamic therapy synergizes with PD-L1 checkpoint blockade for immunotherapy of CRC by multifunctional nanoparticles. Mol. Ther. 2021, 29, 2931–2948. [Google Scholar] [CrossRef]
- Yang, Q.; Guo, N.; Zhou, Y.; Chen, J.; Wei, Q.; Han, M. The role of tumor-associated macrophages (TAMs) in tumor progression and relevant advance in targeted therapy. Acta Pharm. Sin. B 2020, 10, 2156–2170. [Google Scholar] [CrossRef] [PubMed]
- Kumar, A.; Moralès, O.; Mordon, S.; Delhem, N.; Boleslawski, E. Could Photodynamic Therapy Be a Promising Therapeutic Modality in Hepatocellular Carcinoma Patients? A Critical Review of Experimental and Clinical Studies. Cancers 2021, 13, 5176. [Google Scholar] [CrossRef] [PubMed]
- Sun, W.; Cheng, Y.; Ma, X.; Jin, Z.; Zhang, Q.; Wang, G. Photodynamic therapy upregulates expression of HIF-1α and PD-L1 in related pathways and its clinical relevance in non-small-cell lung cancer. Eur. J. Med. Res. 2024, 29, 230. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Lu, Y.; Li, X.; Luo, L.; You, J. Nanoplatform-enhanced photodynamic therapy for the induction of immunogenic cell death. J. Control Release 2024, 365, 1058–1073. [Google Scholar] [CrossRef]
- HIF1A. Wikipedia, The Free Encyclopedia. Available online: https://en.wikipedia.org/wiki/HIF1A (accessed on 31 January 2025).
- Konstantinidou, M.; Li, J.; Zhang, B.; Wang, Z.; Shaabani, S.; Ter Brake, F.; Essa, K.; Dömling, A. PROTACs- a game-changing technology. Expert. Opin. Drug Discov. 2019, 14, 1255–1268. [Google Scholar] [CrossRef]
- Yang, Q.; Zhao, J.; Chen, D.; Wang, Y. E3 ubiquitin ligases: Styles, structures and functions. Mol. Biomed. 2021, 2, 23. [Google Scholar] [CrossRef]
- Li, X.; Pu, W.; Zheng, Q.; Ai, M.; Chen, S.; Peng, Y. Proteolysis-targeting chimeras (PROTACs) in cancer therapy. Mol. Cancer 2022, 21, 99. [Google Scholar] [CrossRef]
- Tong, F.; Wang, Y.; Xu, Y.; Zhou, Y.; He, S.; Du, Y.; Yang, W.; Lei, T.; Song, Y.; Gong, T.; et al. MMP-2-triggered, mitochondria-targeted PROTAC-PDT therapy of breast cancer and brain metastases inhibition. Nat. Commun. 2024, 15, 10382. [Google Scholar] [CrossRef]
- Yu, H.; Chen, B.; Huang, H.; He, Z.; Sun, J.; Wang, G.; Gu, X.; Tang, B.Z. AIE-Active Photosensitizers: Manipulation of Reactive Oxygen Species Generation and Applications in Photodynamic Therapy. Biosensors 2022, 12, 348. [Google Scholar] [CrossRef]
- Li, S.; Liu, Y.; Zhang, X.; Liu, Y.; Si, L.; Jiang, S.; Wang, A.; Che, X.; Chen, J.; Hu, J. Multi-pathway oxidative stress amplification via controllably targeted nanomaterials for photoimmunotherapy of tumors. J. Nanobiotechnol. 2025, 23, 33. [Google Scholar] [CrossRef] [PubMed]
- Duan, Z.; Li, L.; Zhan, Q.; Chen, J.; Li, Q.; Liu, R.; Tu, Y. Mitochondria-Targeting Type-I Photodynamic Therapy Based on Phenothiazine for Realizing Enhanced Immunogenic Cancer Cell Death via Mitochondrial Oxidative Stress. Int. J. Nanomed. 2025, 20, 125–139. [Google Scholar] [CrossRef] [PubMed]
- Tian, J.; Huang, B.; Hasnain Nawaz, M.; Zhang, W. Recent advances of multi-dimensional porphyrin-based functional materials in photodynamic therapy. Coord. Chem. Rev. 2020, 420, 213410. [Google Scholar] [CrossRef]
- Zhao, X.; Liu, J.; Fan, J.; Chao, H.; Peng, X. Recent progress in photosensitizers for overcoming the challenges of photodynamic therapy: From molecular design to application. Chem. Soc. Rev. 2021, 50, 4185–4219. [Google Scholar] [CrossRef]
- Samperi, M.; Limón, D.; Amabilino, D.B.; Pérez García, L. Enhancing Singlet Oxygen Generation by Self-Assembly of a Porphyrin Entrapped in Supramolecular Fibers. Cell Rep. Phys. Sci. 2020, 1, 100030. [Google Scholar] [CrossRef]
- Zhang, P.; Shen, Q.; Yang, J.; Zhao, Z.; Gao, A.; Chen, S.; Zhang, Y.; Meng, L.; Dang, D. Shuttle-like nanoassemblies by isomeric photosensitizers to enhance ROS generation and tumor penetration for photodynamic therapy. Matter 2024, 7, 4342–4355. [Google Scholar] [CrossRef]
- Lee, H.C.; Wei, Y.H. Mitochondrial role in life and death of the cell. J. Biomed. Sci. 2000, 7, 2–15. [Google Scholar] [CrossRef]
- Wang, S.F.; Tseng, L.M.; Lee, H.C. Role of mitochondrial alterations in human cancer progression and cancer immunity. J. Biomed. Sci. 2023, 30, 61. [Google Scholar] [CrossRef]
- Ailioaie, L.M.; Ailioaie, C.; Litscher, G. Gut Microbiota and Mitochondria: Health and Pathophysiological Aspects of Long COVID. Int. J. Mol. Sci. 2023, 24, 17198. [Google Scholar] [CrossRef]
- Antonucci, S.; Di Lisa, F.; Kaludercic, N. Mitochondrial reactive oxygen species in physiology and disease. Cell Calcium. 2021, 94, 102344. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Pan, C.; Feng, C.; Yan, C.; Yu, Y.; Chen, Z.; Guo, C.; Wang, X. Role of mitochondrial reactive oxygen species in homeostasis regulation. Redox Rep. 2022, 27, 45–52. [Google Scholar] [CrossRef] [PubMed]
- Mukherjee, A.; Ghosh, K.K.; Chakrabortty, S.; Gulyás, B.; Padmanabhan, P.; Ball, W.B. Mitochondrial Reactive Oxygen Species in Infection and Immunity. Biomolecules 2024, 14, 670. [Google Scholar] [CrossRef]
- Li, H.; Shen, J.; Zheng, C.; Zhu, P.; Yang, H.; Huang, Y.; Mao, X.; Yang, Z.; Hu, G.; Chen, Y. Cell death: The underlying mechanisms of photodynamic therapy for skin diseases. Interdiscip. Med. 2025, e20240057. [Google Scholar] [CrossRef]
Reference | Type of Study | Nanotechnology Tested | Results | Conclusions |
---|---|---|---|---|
Mao, D. et al., 2017, [24] | 4T1 breast cancer cells were inoculated subcutaneously into normal BALB/c mice, resulting in a mammary tumor-bearing mouse model. | In vivo chemiluminescence of newly designed C-TBD NPs with 1O2 production. | C-TBD NPs showed high FR/NIR chemiluminescence and powerful singlet production in both primary and peritoneal metastatic mouse models. An innovative blueprint for simultaneous tumor diagnosis and treatment. | Selective chemiexcited image-guided and light-source-free PDT for deep tumors. Original platform for accurate cancer theranostics. |
Li, M. et al., 2018, [25] | In vitro and in vivo models for PDT against hypoxic tumors. HepG2 or COS-7 cells, incubated on cell culture plates. Subcutaneous tumor model and in vivo cancer imaging in 6–8-week-old female Balb/c mice, subcutaneously injected with 1 × 106 H22 cells to establish a liver tumor model. | Near-Infrared Light-Initiated Molecular Superoxide Radical Generator (ENBS-B with biotin ligand). ENBS-B or ENBS-C6-NH2 was injected intravenously into tumor bearing mice. | ENBS-B reaches 87- times higher cellular uptake in cancer cells than in normal ones. New options for personalized medicine and exceptional agents for clinical cancer therapy. | ENBS-B accurately targets neoplastic network cells and fully eliminates tumor growth upon low-dose light- irradiation. |
Sun, CL. et al., 2019, [26] | In vitro Ald-DSB as 2PEF imaging on MCF-7 human breast cancer cells; and in vivo turn-on 2PE-PDT in a mice tumor model of melanoma. BALB/c nude mice were inoculated subcutaneously with B16-F10 melanoma cells. | In vitro 2PE-PDT on MCF-7 human breast cancer cells; laser irradiation at 760 nm (10 mW/cm2 120 fs, 1000 Hz) for 20 min. For in vivo imaging and 2PA-PDT, mice were injected with Ace-DSB at the tumor location. Light treatment of 80 mW/cm2 was combined with Ace-DSB. 2PE-PDT was performed for 10 min, three times. | A simulation-assisted strategy for fast screening of small molecules to design turn-on-type 2PA dyes for simultaneous imaging and PDT of neoplasms. By turn-on only at tumor site, the phototoxicity to healthy cells can be decreased. | Practical implementation of the first screening for new theranostic chemical substances. |
Guan, Q. et al., 2019, [27] | PDT in vitro and in vivo experiments on HeLa and MCF-7 cancer cell lines. Nude mice (BALB/c-nu♀, aged 5 weeks, 15~20 g) were subcutaneously injected MCF-7 cancer cells (5 × 106 cells), to set up the MCF-7 xenograft model. | NCOF-based PSs for PDT: two BODIPY-decorated NCOFs and two amino-decorated BODIPY molecules. Green LED irradiation (40 mW/cm2), for different times: 0 (control) to 15 min. | PDT in vitro of nanoscale LZU-1-BODIPY-2I and LZU-1-BODIPY-2H, followed by high-efficacy PDT in vivo: successfully inhibited the MCF-7 xenograft growth without systemic toxicity, localized to lysosomes and mitochondria; cell death through mitochondrion-lysosome-connected pathways. Intracellular 1O2 imaging, cytostatic and other tests proved that both NCOF-based PSs are excellent nanomedical agents for PDT, especially the first one, with heavy atoms of iodine. | NCOF-based PSs tested for PDT proved to be high-performing agents, due to low cytotoxicity, fine biocompatibility, high uptake by cancer cells and highly efficient 1O2 generation. BDF procedure is yielding and many-sided for state-of-the art NCOF-based PDT materials, by covalently transfer of bioactive, chemotherapeutic, and targeting agents for clinical cancer treatment. |
Wu, Q. et al., 2019, [28] | In vitro BPN/MnO2 -catalyzed 1O2 production within HeLa cells. In vivo efficacy against mice bearing HeLa tumors. In vivo antitumor effects of BPN/MnO2/DOX, followed by 660 nm photoirradiation and/or NIR 808 nm laser irradiation (0.8 W/cm2, 5 min). | Smart BPN/MnO2—tumor-driven nanotheranostic platform for T1-weighted MRI-guided synergistic PDT, PTT, and chemotherapy | Efficient 1O2 generation inside cancer cells under laser irradiation (660 nm, 0.22 W/cm2, 10 min). Significantly enhanced PDT and PTT performance. Outstanding antitumor performance of BPN/MnO2/DOX-based multimodality therapy. | Facile synthesis of compact BPN/MnO2 nanohybrid platform, with following advantages: -finite systemic cytotoxicity; -reduced therapeutic resistance; -precise spatial selectivity; -easy operation. Multifunctional BPN/MnO2/DOX inhibited tumor growth without relapses. |
Wei, S.M. et al., 2020, [29] | In vitro PDT for CDs on tumor cell lines of human hepatoma Bel-7402, human lung adenocarcinoma A549, human cervical carcinoma HeLa, as well as normal epithelial cells of L929. In vivo PDT for CDs: intravenously administered tumor-bearing mice. Irradiation with laser (589 nm, 0.5 W/cm2). | Quantitative analysis of intracellular O2′− levels with different CDs. In vivo fluorescence imaging: maximum fluorescence on tumor tissue occurred 4 h after injection. | Large-scale karyopyknosis and necrosis in tumor tissues with CD and laser irradiation. No significant morphological changes or side effects in vital organs. | Extensive generation of luminescent CDs exhibiting an outstanding power to photogenerate O2′− with pronounced photocytotoxicity. Exploratory implementations of carbon-based materials in PDT and photocatalytic reactions. |
Wang, D. et al., 2020, [30] | In vitro PDT efficacy and biocompatibility of OxgeMCC-r SAE against 4T1 tumor cells. In vivo PDT on 4T1 tumor- bearing mice, divided into 4 groups (control, free Ce6, MCC, and OxgeMCC-r). Laser irradiation (671 nm laser, 100 mWcm−2, 5 min) at 6 h post intravenous injection. | OxgeMCC-r SAE followed in vivo MRI-guided tumor PDT. | Remarkable accumulation of OxgeMCC-r inside the tumor 6 h after injection. Shrinkage of tumor on MRI 24 h after injection and PDT. Approx. total tumor elimination 3 days after PDT. | OxgeMCC-r SAE, a prospective theranostic nanoplatform for MRI-guided cancer therapy. |
Zheng, B. et al., 2021, [31] | In vitro and in vivo PDT for treating deep-tumor models at low laser irradiation (660 nm and 808 nm) on CT26 cells, SKOV3 human ovarian cancer cells; and SKOV3-tumor bearing mice, intravenously injected with 200 mL of the nanoconjugates (10 mg/mL). In vivo tumor site fluorescence intensity and T1 -weighted MRI 24 h post-injection of NaGdF4:Nd-Ce6 nanoconjugates. Mice were treated with an 808 nm laser for 30 min, at 80 mW/cm2. | NaGdF4:Nd-Ce6 as a PDT agent. | Avoiding intersystem crossing by applying inorganic lanthanide nanocrystals, which act directly on the triplet excited states of PS. Greater penetration depth of 808 nm light in solid tumors. Much more apoptotic cells in histological sections from the NaGdF4:Nd-Ce6-FA+ 808 nm laser group. | A completely new design for lanthanide-triplet NIR photosensitization for increased ROS in PDT. Decreased energy losses. Much increased efficacy for deep cancer treatments at ultralow irradiation. |
Yao, Q. et al., 2022, [32] | In vitro and in vivo PDT on breast cancer cells (MCF-7) and MCF-7-adr, i.e., resistant adriamycin breast cancer cells. Irradiated for 10 min (660 nm, 15 mW/cm2). In vivo PDT of multiple tumor models. Tumor-bearing mice were treated with light irradiation (50 mW/cm2 for 10 min) | NBEX (X = S, Se, Te), as PS with a type-III mechanism. | NBEX quickly changes to NP in a hydrophilic environment. Very good tumor elimination action for various tumors (subcutaneous, primary or glioma) and inhibition of metastases. | NBEX, as an oxygen-independent PS, immediately transfers light energy to the RNA of neoplastic cells and kills them with exceptional efficiency, also stimulating host immune responses to fight metastases. |
Lu, B. et al., 2024, [37] | In vitro PDT towards NIH 3T3 cells, and tumor HeLa cells. | 3 novel PSs: BODIPY-CD complexes tested for concentrations from 12.5 to 200 μg/mL. | Maximum emission peaks for alkynyl-BODIPY dye and BODIPY-CD complex at 783 nm and 874 nm, respectively. BODIPY-β-CD had a faster and higher ROS formation rate compared to the other two. HeLa cells viability decreased to 20% (at 100 μg/mL) with laser irradiation. | BODIPY-β-CD complex had best PDT activity against HeLa cells. |
Li, L. et al., 2024, [44] | In vitro experiments on U87 -human glioblastoma cells and on U-CH1 -human chordoma cell line. In vivo murine model of brain tumor xenograft. 2.0 W NIR excitation power, 20 min of irradiation time. | NO-releasing platform for UCNPs@mSiO2-NH2&RBS | In vitro NO set free after NIR illumination of the platform restricted migration, reduced cells activity to 50% and triggered necrotic and apoptotic effects on neoplastic cell lines. In vivo study showed a significant halt in tumor growth in the treated group of mice, without adverse reactions. | Recently developed photocontrolled NO release platform by NIR photoexcitation of UCNPs@mSiO2-NH2&RBS may constitute an effective and safe therapy option for solid tumors. |
Qiao, Y. et al., 2024, [50] | In vitro PDT on murine breast cancer 4T1 cells (4T1). Hemocompatibility studied in red blood cells. | Multifunctional CAT@MnPFs with nanometric size, i.e., ~114 nm. | CAT@MnPFs display a top 1O2 production. CAT@MnPFs killed 4T1 cells upon 650 nm irradiation (100 mW/cm2) for 10 min at a rate of 80.1% in the H2O2 group. High biosafety, negligible hemolysis at even 300 μg mL−1. | CAT@MnPFs, can give rise to O2 for pushing upward PDT. |
References | Type of Study | Photosensitizer | Forms of Cancer | Effects on Mitochondria |
---|---|---|---|---|
Yang, Z. et al., 2019, [52] | In vitro and in vivo experiments | Mn3O4@MSNs@IR780 nanoparticles | Human gastric cancer cell line MKN-45P and tumor-bearing MKN-45P xenograft mice | Mn3O4@MSNs@IR780 can prevent tumor hypoxia in vivo. They offer highly targeted qualities towards mitochondria, with the ability to sustainably inhibit tumor hypoxia. |
Wen, J. et al., 2021, [53] | In vitro and in vivo experiments | Multifunctional 3BP@PLGA-IR780 nanoplatform | Murine breast cancer line 4T1 cells and 4T1 tumor-bearing mice models | 3BP@PLGA-IR780 nanoplatform penetrates deeply into the tumor, reduces tumor cells’ oxygen levels, enhances ROS generation, precisely targets and destroys tumor cell mitochondria. |
Nash, G.T. et al., 2021, [54] | In vitro and in vivo experiments | New nanoscale metal–organic layer (nMOL) assembly, ZnOPPc@nMOL, with ZnOPPc [ZnOPPc = zinc(II)-2,3,9,10,16,17,23,24-octa(4-carboxyphenyl phthalocyanine] or ZnOPPc@nMOL | MC38 tumor-bearing C57BL/6 mice and CT26 tumor-bearing BALB/c mice | PDT with ZnOPPc@nMOL demonstrated mitochondrial penetration and >99% tumor growth inhibition efficiency, and 40–60% cure rates on 2 mouse models of colon cancer. |
Cai, X. et al., 2021, [55] | In vitro and in vivo experiments | Multifunctional Ir-NPs nano-complex | SKOV3 ovarian carcinoma cell line and SKOV3 tumor-bearing mice | In vitro and in vivo Ir-NPs administered in PDT penetrated deeply into the tumor, disrupted mitochondrial redox homeostasis and induced apoptosis. |
Huang, Z. et al., 2022, [59] | In vitro experiments | Amphiphilic heterodimeric photosensitizer (TPP-TK-PPa) and TPP-TK-PPa/DEM NPs nanoplatform | MDA-MB-231 breast cancer cell line as “triple-negative” breast cancer | TPP-TK-PPa/DEM NPs applied in PDT produced remarkable cytotoxicity in MDA-MB-231 cell line and may provide precise and efficient targeting capability on tumor cells. |
Bonelli, J. et al., 2022, [63] | In vitro experiments | Coumarin PS in amphoteric polyurethane-polyurea hybrid nanocapsules. Nanoformulation NC-COUPY 2 | Human cervix adenocarcinoma cell line. HeLa, buffalo green monkey kidney cells, BGM and MTCS | In vitro NC-COUPY 2 experimentally PDT demonstrated potent tumor growth inhibition effect, high phototoxic profile, mitochondrial degradation through autophagy, and apoptosis of cancer cells compared to free coumarin. |
Li, X. et al., 2022, [74] | In vitro and in vivo experiments | RBCm@ATO-IR780-PFC liposomes | Human gastric cancer line AGS and in vivo, mice with tumors induced by CT26 colorectal cancer cells | Biomimetic nanoparticles with ATO under the action of PDT reversed hypoxia in vitro and in vivo and had prominent antitumor action on targeted mitochondria. |
Zhao, X. et al., 2024, [79] | In vitro and in vivo study | Theranostic agents (TPAPyTZ, TPAPyTC, TPAPyTM, and TPAPyTI | HCT116 and HT29 colorectal cancer cells and HCT116-tumor bearing mice | TPAPyTZ induced GA-ROS, leading to GA breakdown, and activation of mitochondria caspase-related apoptosis, i.e., vigorous apoptosis of cancer cells. |
Chen, M. et al., 2024, [86] | In vitro and in vivo study | Multifunctional DNA nanoclew-AS-AMD | Study on 4T1 breast tumor cells and 4T1-tumor- bearing mice | The intelligent poly aptamer-encoded DNA nanoclew activated mitochondria-targeted PDT and MRI. The AS-AMD platform substantially improved MRI performance and PDT efficacy. |
Zhou, Y. et al., 2024, [92] | In vitro and in vivo study | IR700DX-6T-PDT | Human CRC cell lines, Murine colon adenocarcinoma cell line (CT26), HCT11 CRC cells, and BALB/c mouse model with CT26 tumor | IR700DX-6T-PDT induced pyroptosis in CRC in experimental mice via the ROS/p38/CASP3/GSDME axis and in this way could activate microsatellite stable colorectal cancer (MSS-CRC) to PD-1 blockade therapy. |
Li, J. et al., 2024, [96] | In vitro and in vivo study | EHMONs-Ce6-CTPP@PFC nanoplatform | 4T1 human breast cancer cells and 4T1 breast tumor model by BALB/c mice | EHMONs-CTPP-Ce6@PFC nanoplatform has good biocompatibility and enhanced efficiency in targeting mitochondria, alleviating hypoxia and generating singlet oxygen. |
Liu, Z.H. et al., 2024, [97] | In vitro study | CMPNs photodynamic nanoplatform targeting CD44 and mitochondria | Human lung adenocarcinoma cell line A549 | FRET mediation and mitochondria targeting with no cytotoxicity from CMPN-treated A549 cells. CD44 and mitochondria-targeted nanoplatform improved 1O2 production, mitochondrial targeting, mitochondrial depolarization and FRET-mediated PDT in CMPNs is a powerful master plan for enhanced PDT. |
Tong, F. et al., 2024, [107] | In vitro and in vivo study | Multifunctional PROTAC-PDT nanoplatform (dBET6@CFMPD) | Murine 4T1 breast cancer cells, and 4T1-Luc cells Murine E0771 breast cancer cells and E0771-Luc cells. Brain-metastasis model of breast cancer 4 T1-Luc cells or E0771 Luc cells | dBET6@CFMPD platform targeting mitochondria in vitro and in vivo confirmed that it can stop breast cancer progression and its metastases through the association of PROTAC-PDT and TIME remodeling. |
Duan, Z. et al., 2025, [110] | In vitro and in vivo study | Rh-PTZ NPs type I photosensitizer targeting mitochondria | 4T1 cancer cell lines and 4T1 tumor carriers by female BALB/c mice | Rh-PTZ administered in PDT on 4T1 cancer cells lines and 4T1 tumor-bearing mice, confirmed mitochondrial targeting, ROS generation, ICD activation, apoptosis and necrosis of tumor cells. |
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ailioaie, L.M.; Ailioaie, C.; Litscher, G. Fighting Cancer with Photodynamic Therapy and Nanotechnologies: Current Challenges and Future Directions. Int. J. Mol. Sci. 2025, 26, 2969. https://doi.org/10.3390/ijms26072969
Ailioaie LM, Ailioaie C, Litscher G. Fighting Cancer with Photodynamic Therapy and Nanotechnologies: Current Challenges and Future Directions. International Journal of Molecular Sciences. 2025; 26(7):2969. https://doi.org/10.3390/ijms26072969
Chicago/Turabian StyleAilioaie, Laura Marinela, Constantin Ailioaie, and Gerhard Litscher. 2025. "Fighting Cancer with Photodynamic Therapy and Nanotechnologies: Current Challenges and Future Directions" International Journal of Molecular Sciences 26, no. 7: 2969. https://doi.org/10.3390/ijms26072969
APA StyleAilioaie, L. M., Ailioaie, C., & Litscher, G. (2025). Fighting Cancer with Photodynamic Therapy and Nanotechnologies: Current Challenges and Future Directions. International Journal of Molecular Sciences, 26(7), 2969. https://doi.org/10.3390/ijms26072969