Unleashing the power of porphyrin photosensitizers: Illuminating breakthroughs in photodynamic therapy

Alibasha Akbar1, Syamantak Khan2, Tanmay Chatterjee3, Mihir Ghosh1
1Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
2Department of Radiation Oncology, Stanford University, Stanford, CA, USA
3Department of Chemistry, Birla Institute of Technology & Science, Pilani Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India

Tài liệu tham khảo

Liu, 2021, Nanoparticles loading porphyrin sensitizers in improvement of photodynamic therapy for ovarian cancer, Photodiagn. Photodyn. Ther., 33, 102156, 10.1016/j.pdpdt.2020.102156 Karunakaran, 2013, In vitro demonstration of apoptosis mediated photodynamic activity and NIR nucleus imaging through a novel porphyrin, ACS Chem. Biol., 8, 127, 10.1021/cb3004622 Hu, 2015, Nanocomposite-based photodynamic therapy strategies for deep tumor treatment, Small, 11, 5860, 10.1002/smll.201501923 Tsolekile, 2019, Porphyrin as diagnostic and therapeutic agent, Molecules, 24, 2669, 10.3390/molecules24142669 Calixto, 2016, Nanotechnology-based drug delivery systems for photodynamic therapy of cancer: a review, Molecules, 21, 342, 10.3390/molecules21030342 Prasanth, 2014, Antimicrobial Photodynamic Efficiency of Novel Cationic Porphyrins towards Periodontal Gram-positive and Gram-negative Pathogenic Bacteria, Photochem. Photobiol., 90, 628, 10.1111/php.12198 Zhou, 2016, Reactive oxygen species generating systems meeting challenges of photodynamic cancer therapy, Chem. Soc. Rev., 45, 6597, 10.1039/C6CS00271D Ethirajan, 2011, The role of porphyrin chemistry in tumor imaging and photodynamic therapy, Chem. Soc. Rev., 40, 340, 10.1039/B915149B Fan, 2016, Overcoming the Achilles' heel of photodynamic therapy, Chem. Soc. Rev., 45, 6488, 10.1039/C6CS00616G Li, 2019, Phthalocyanines as medicinal photosensitizers: Developments in the last five years, Coord. Chem. Rev., 379, 147, 10.1016/j.ccr.2017.08.003 Jayaram, 2016, In vitro and in vivo demonstration of photodynamic activity and cytoplasm imaging through TPE nanoparticles, ACS Chem. Biol., 11, 104, 10.1021/acschembio.5b00537 Moreira, 2008, Photodynamic therapy: porphyrins and phthalocyanines as photosensitizers, Aust. J. Chem., 61, 741, 10.1071/CH08145 Ferreira, 2004, Analysis of mitochondria, endoplasmic reticulum and actin filaments after PDT with AlPcS 4, Lasers Med. Sci., 18, 207, 10.1007/s10103-003-0282-6 Marydasan, 2019, Picolyl porphyrin nanostructures as a functional drug entrant for photodynamic therapy in human breast cancers, Acs Omega, 4, 12808, 10.1021/acsomega.9b01380 Hiroto, 2017, Synthesis and functionalization of porphyrins through organometallic methodologies, Chem. Rev., 117, 2910, 10.1021/acs.chemrev.6b00427 Tanaka, 2015, Conjugated porphyrin arrays: synthesis, properties and applications for functional materials, Chem. Soc. Rev., 44, 943, 10.1039/C3CS60443H Marbach, 2015, Surface-mediated in situ metalation of porphyrins at the solid–vacuum interface, Acc. Chem. Res., 48, 2649, 10.1021/acs.accounts.5b00243 Urbani, 2014, Meso-substituted porphyrins for dye-sensitized solar cells, Chem. Rev., 114, 12330, 10.1021/cr5001964 Shi, 2021, Porphyrin-based compounds and their applications in materials and medicine, Dyes Pigments, 188, 109136, 10.1016/j.dyepig.2021.109136 Gottfried, 2015, Surface chemistry of porphyrins and phthalocyanines, Surf. Sci. Rep., 70, 259, 10.1016/j.surfrep.2015.04.001 Lu, 2011, Catalytic C–H functionalization by metalloporphyrins: recent developments and future directions, Chem. Soc. Rev., 40, 1899, 10.1039/C0CS00070A Lu, 2016, Optically active porphyrin and phthalocyanine systems, Chem. Rev., 116, 6184, 10.1021/acs.chemrev.5b00588 Singh, 2015, Glycosylated porphyrins, phthalocyanines, and other porphyrinoids for diagnostics and therapeutics, Chem. Rev., 115, 10261, 10.1021/acs.chemrev.5b00244 Beletskaya, 2009, Supramolecular chemistry of metalloporphyrins, Chem. Rev., 109, 1659, 10.1021/cr800247a Hembury, 2008, Chirality-sensing supramolecular systems, Chem. Rev., 108, 1, 10.1021/cr050005k Almeida-Marrero, 2018, Porphyrinoid biohybrid materials as an emerging toolbox for biomedical light management, Chem. Soc. Rev., 47, 7369, 10.1039/C7CS00554G Pereira, 2016, Porphyrin-based metal-organic frameworks as heterogeneous catalysts in oxidation reactions, Molecules, 21, 1348, 10.3390/molecules21101348 Borovkov, 2014, Supramolecular chirality in porphyrin chemistry, Symmetry, 6, 256, 10.3390/sym6020256 Ghosh, 2014, Photoinduced electron transfer in metallo-octaethylporphyrin (donor)–2-nitrofluorene (acceptor) systems in polar acetonitrile liquid medium, J. Photochem. Photobiol. A Chem., 290, 94, 10.1016/j.jphotochem.2014.06.013 Ghosh, 2015, Photoinduced electron transfer from zinc tetraphenylporphyrin to 2-nitrofluorene in polar solvent acetonitrile, J. Photochem. Photobiol. A Chem., 306, 55, 10.1016/j.jphotochem.2015.03.019 Ghosh, 2015, Fluorescence quenching of 9-cyanoanthracene by metallo-octaethylporphyrins in cyanobenzene, J. Porphyrins Phthalocyanines, 19, 1063, 10.1142/S1088424615500819 Ghosh, 2015, Solvatochromic Stokes shift and determination of excited state dipole moments of free base and zinc octaethylporphyrin, Spectrochim. Acta A Mol. Biomol. Spectrosc., 150, 959, 10.1016/j.saa.2015.06.057 Ghosh, 2019, Photoinduced electron transfer in non-covalent free-base octaethylporphyrin and 2-nitrofluorene donor-acceptor system: A combined experimental and quantum chemical study, J. Mol. Liq., 277, 656, 10.1016/j.molliq.2018.12.155 Mfouo-Tynga, 2021, Features of third generation photosensitizers used in anticancer photodynamic therapy, Photodiagn. Photodyn. Ther., 34, 102091, 10.1016/j.pdpdt.2020.102091 Kou, 2017, Porphyrin photosensitizers in photodynamic therapy and its applications, Oncotarget, 8, 81591, 10.18632/oncotarget.20189 Tian, 2020, Recent advances of multi-dimensional porphyrin-based functional materials in photodynamic therapy, Coord. Chem. Rev., 420, 213410, 10.1016/j.ccr.2020.213410 Rabiee, 2020, Recent advances in porphyrin-based nanocomposites for effective targeted imaging and therapy, Biomaterials, 232, 119707, 10.1016/j.biomaterials.2019.119707 Detty, 2004, Current clinical and preclinical photosensitizers for use in photodynamic therapy, J. Med. Chem., 47, 3897, 10.1021/jm040074b Ramakrishnan, 2012 Allison, 2010, Oncologic photodynamic therapy photosensitizers: a clinical review, Photodiagn. Photodyn. Ther., 7, 61, 10.1016/j.pdpdt.2010.02.001 Zhang, 2018, An updated overview on the development of new photosensitizers for anticancer photodynamic therapy, Acta Pharm. Sin. B, 8, 137, 10.1016/j.apsb.2017.09.003 Sharman, 1999, Photodynamic therapeutics: basic principles and clinical applications, Drug Discov. Today, 4, 507, 10.1016/S1359-6446(99)01412-9 Nyman, 2004, Research advances in the use of tetrapyrrolic photosensitizers for photodynamic therapy, J. Photochem. Photobiol. B Biol., 73, 1, 10.1016/j.jphotobiol.2003.10.002 Sternberg, 1998, Porphyrin-based photosensitizers for use in photodynamic therapy, Tetrahedron, 54, 4151, 10.1016/S0040-4020(98)00015-5 Pushpan, 2002, Porphyrins in photodynamic therapy-a search for ideal photosensitizers, Curr. Med. Chem.-Anti-Cancer Agents, 2, 187, 10.2174/1568011023354137 Kudinova, 2010, Photodynamic therapy of cancer: Search for ideal photosensitizer, Biochem. (Moscow) Suppl. Series B: Biomedi. Chem., 4, 95, 10.1134/S1990750810010129 DeRosa, 2002, Photosensitized singlet oxygen and its applications, Coord. Chem. Rev., 233, 351, 10.1016/S0010-8545(02)00034-6 Wang, 2019, Type I photodynamic therapy by organic–inorganic hybrid materials: From strategies to applications, Coord. Chem. Rev., 395, 46, 10.1016/j.ccr.2019.05.016 Cunningham, 1966, Experimental evaluation of hematoporphyrin in the detection and management of intraocular tumors, Am J. Ophthalmol., 61, 36, 10.1016/0002-9394(66)90744-6 Thaller, 1983, Potential use of radiolabelled porphyrins for tumor scanning, Porphyr. Photosensitizat., 265, 10.1007/978-1-4684-4406-3_23 Goldman, 2013 Kömerik, 2002, A novel approach to cancer treatment: Photodynamic therapy, Turk. J. Cancer, 32, 83 Schmidt-Erfurth, 1998, Photodynamic therapy of subfoveal choroidal neovascularization: clinical and angiographic examples, Graefes Arch. Clin. Exp. Ophthalmol., 236, 365, 10.1007/s004170050092 Jin, 1990, Review of photodynamic therapy for gastrointestinal tumours in the past 6 years in China, J. Photochem. Photobiol. B, 7, 87, 10.1016/1011-1344(90)85145-M Stylli, 2004, Photodynamic therapy of brain tumours: evaluation of porphyrin uptake versus clinical outcome, J. Clin. Neurosci., 11, 584, 10.1016/j.jocn.2004.02.001 Stylli, 2005, Photodynamic therapy of high grade glioma–long term survival, J. Clin. Neurosci., 12, 389, 10.1016/j.jocn.2005.01.006 Clarke, 2006, Management of malignant mesothelioma by decortication and adjunct phototherapy, Asian Cardiovasc. Thorac. Ann., 14, 206, 10.1177/021849230601400307 Peterson, 1992, Photodynamic therapy of human ovarian epithelial carcinoma, OVCAR-3, heterotransplanted in the nude mouse, Am. J. Obstet. Gynecol., 167, 1852, 10.1016/0002-9378(92)91786-A Nauta, 1997, Photofrin-mediated photodynamic therapy of chemically-induced premalignant lesions and squamous cell carcinoma of the palatal mucosa in rats, Int. J. Oral Maxillofac. Surg., 26, 223, 10.1016/S0901-5027(97)80825-9 Wang, 2015, Analysis of the in vivo and in vitro effects of photodynamic therapy on breast cancer by using a sensitizer, sinoporphyrin sodium, Theranostics, 5, 772, 10.7150/thno.10853 Sun, 2016, Curative effect of the recent photofrin photodynamic adjuvant treatment on young patients with advanced colorectal cancer, Oncol. Lett., 11, 2071, 10.3892/ol.2016.4179 Qiu, 2017, A Comparison of Dose Metrics to Predict Local Tumor Control for Photofrin-mediated Photodynamic Therapy, Photochem. Photobiol., 93, 1115, 10.1111/php.12719 Oseroff, 2006, A dose ranging study of photodynamic therapy with porfimer sodium (Photofrin) for treatment of basal cell carcinoma, Lasers Surg. Med., 38, 417, 10.1002/lsm.20363 Porter, 2014, Evidence and conjecture about mechanisms of cutaneous disease in photodermatology, Exp. Dermatol., 23, 543, 10.1111/exd.12467 Cai, 2013, Photodynamic therapy for facial actinic keratosis: a clinical and histological study in Chinese patients, Photodiagn. Photodyn. Ther., 10, 260, 10.1016/j.pdpdt.2012.12.003 Millard, 2002, Photosensitivity disorders: cause, effect and management, Am. J. Clin. Dermatol., 3, 239, 10.2165/00128071-200203040-00002 El-Far, 1988, Selectivein vivo tumor localization of heptacar☐ ylic porphyrin isomer I in a bladder tumor model: a novel technique to modulate porphyrin localization, Biochimie, 70, 1379, 10.1016/0300-9084(88)90010-7 Moan, 1987, Photosensitizing efficiencies, tumor-and cellular uptake of different photosensitizing drugs relevant for photodynamic therapy of cancer: section IV–Cellular Photosensitization, Photochem. Photobiol., 46, 713, 10.1111/j.1751-1097.1987.tb04837.x Christensen, 1981, Photodynamic effects of haematoporphyrin derivative on synchronized and asynchronous cells of different origin, Br. J. Cancer, 44, 717, 10.1038/bjc.1981.258 Marko, 2016, Multifunctional Agents for Cancer-Imaging and Photodynamic Therapy: Impact of Polyacrylamide-Based Nanoplatforms, Handbook of photodynamic therapy: Updates on recent applications of porphyrin-based compounds, World Sci., 3 Tang, 2005, Photodynamic characterization and in vitro application of methylene blue-containing nanoparticle platforms, Photochem. Photobiol., 81, 242 Zhang, 2018, An updated overview on the development of new photosensitizers for anticancer photodynamic therapy, Acta Pharm. Sin. B, 8, 137, 10.1016/j.apsb.2017.09.003 Baskaran, 2018, Clinical development of photodynamic agents and therapeutic applications, Biomater. Res., 22, 25, 10.1186/s40824-018-0140-z Yoon, 2013, Advance in photosensitizers and light delivery for photodynamic therapy, Clin. Endosc., 46, 7, 10.5946/ce.2013.46.1.7 Moriwaki, 2018, Synthesis and photophysical properties of S-mannosylated chlorins and their effect on photocytotoxicity in HeLa cells, Bull. Chem. Soc. Jpn., 91, 230, 10.1246/bcsj.20170271 Wall, 1997, Photoinduced electron transfer in covalently linked oxomolybdenum (V) porphyrin systems, Inorg. Chem., 36, 5676, 10.1021/ic9705711 Yedukondalu, 2011, Core-modified porphyrin based assemblies, Coord. Chem. Rev., 255, 547, 10.1016/j.ccr.2010.10.039 Josefsen, 2008, Photodynamic therapy: novel third-generation photosensitizers one step closer?, Br. J. Pharmacol., 154, 1, 10.1038/bjp.2008.98 Wall, 1997, Photoinduced Electron Transfer in Covalently Linked Oxomolybdenum(V) Porphyrin Systems, Inorg. Chem., 36, 5676, 10.1021/ic9705711 Magda, 2006, 466 Young, 1996, Lutetium texaphyrin (PCI-0123): a near-infrared, water-soluble photosensitizer, Photochem. Photobiol., 63, 892, 10.1111/j.1751-1097.1996.tb09647.x Forsyth, 1995, Synthesis and structure of a 5, 15-bis (4-pyridyl) purpurin, Tetrahedron Lett., 36, 9093, 10.1016/0040-4039(95)01963-I Ryu, 2021, Interrogating biological systems using visible-light-powered catalysis, Nat. Rev. Chem., 5, 322, 10.1038/s41570-021-00265-6 Guardiano, 1989, Tetra-n-propylporphycene as a tumour localizer: pharmacokinetic and phototherapeutic studies in mice, Cancer Lett., 44, 1, 10.1016/0304-3835(89)90100-6 Taneja, 2016, Final Results of a Phase I/II Multicenter Trial of WST11 Vascular Targeted Photodynamic Therapy for Hemi-Ablation of the Prostate in Men with Unilateral Low Risk Prostate Cancer Performed in the United States, J. Urol., 196, 1096, 10.1016/j.juro.2016.05.113 Tardivo, 2005, Methylene blue in photodynamic therapy: From basic mechanisms to clinical applications, Photodiagn. Photodyn. Ther., 2, 175, 10.1016/S1572-1000(05)00097-9 Graciano, 2015, Using chitosan gels as a toluidine blue O delivery system for photodynamic therapy of buccal cancer: in vitro and in vivo studies, Photodiagn. Photodyn. Ther., 12, 98, 10.1016/j.pdpdt.2014.11.003 Cincotta, 1993, Phototoxicity, redox behavior, and pharmacokinetics of benzophenoxazine analogues in EMT-6 murine sarcoma cells, Cancer Res., 53, 2571 Harris, 2004, An investigation into the potential of phenothiazinium-based photo-sensitisers to act as PDT agents, Photodiagn. Photodyn. Ther., 1, 231, 10.1016/S1572-1000(04)00046-8 Harris, 2005, Phenothiazinium based photosensitisers--photodynamic agents with a multiplicity of cellular targets and clinical applications, Curr. Drug Targets, 6, 615, 10.2174/1389450054545962 Kennedy, 1990, Photodynamic therapy with endogenous protoporphyrin IX: basic principles and present clinical experience, J. Photochem. Photobiol. B, 6, 143, 10.1016/1011-1344(90)85083-9 Ormond, 2013, Dye Sensitizers for Photodynamic Therapy, Materials (Basel), 6, 817, 10.3390/ma6030817 Biel, 2006, Advances in photodynamic therapy for the treatment of head and neck cancers, Lasers Surg. Med., 38, 349, 10.1002/lsm.20368 Biel, 2010, 281 Vogel, 1986, Porphycene—a novel porphin isomer, Angew. Chem. Int. Ed. Engl., 25, 257, 10.1002/anie.198602571 Anguera, 2017, Porphycenes and Related Isomers: Synthetic Aspects, Chem. Rev., 117, 2481, 10.1021/acs.chemrev.6b00345 de Moraes, 2015, Effects of photodynamic therapy mediated by nanoemulsion containing chloro-aluminum phthalocyanine: a histologic and immunohistochemical study in human gingiva, Photodiagn. Photodyn. Ther., 12, 592, 10.1016/j.pdpdt.2015.10.009 Wainwright, 2008, Photodynamic therapy: the development of new photosensitisers, Anti Cancer Agents Med. Chem., 8, 280, 10.2174/187152008783961888 Borgatti-Jeffreys, 2005, 624 Miller, 2007, Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: the case experience with preclinical mechanistic and early clinical-translational studies, Toxicol. Appl. Pharmacol., 224, 290, 10.1016/j.taap.2007.01.025 Muehlmann, 2015, Aluminium-phthalocyanine chloride nanoemulsions for anticancer photodynamic therapy: Development and in vitro activity against monolayers and spheroids of human mammary adenocarcinoma MCF-7 cells, J. Nanobiotechnol., 13, 36, 10.1186/s12951-015-0095-3 Delaey, 2000, A comparative study of the photosensitizing characteristics of some cyanine dyes, J. Photochem. Photobiol. B Biol., 55, 27, 10.1016/S1011-1344(00)00021-X Panzarini, 2014, Rose Bengal acetate photodynamic therapy (RBAc-PDT) induces exposure and release of Damage-Associated Molecular Patterns (DAMPs) in human HeLa cells, PLoS One, 9, 10.1371/journal.pone.0105778 Panzarini, 2011, Timing the multiple cell death pathways initiated by Rose Bengal acetate photodynamic therapy, Cell Death Dis., 2, 10.1038/cddis.2011.51 Dini, 2010, Rose bengal acetate photodynamic therapy-induced autophagy, Cancer Biol. Ther., 10, 1048, 10.4161/cbt.10.10.13371 Garg, 2012, Hypericin-based photodynamic therapy induces surface exposure of damage-associated molecular patterns like HSP70 and calreticulin, Cancer Immunol. Immunother., 61, 215, 10.1007/s00262-011-1184-2 Kitanov, 2001, Hypericin and pseudohypericin in some Hypericum species, Biochem. Syst. Ecol., 29, 171, 10.1016/S0305-1978(00)00032-6 Kubin, 2005, Hypericin-the facts about a controversial agent, Curr. Pharm. Des., 11, 233, 10.2174/1381612053382287 Lim, 2010, In vitro and in vivo photocytotoxicity of boron dipyrromethene derivatives for photodynamic therapy, J. Med. Chem., 53, 2865, 10.1021/jm901823u Awuah, 2012, Boron dipyrromethene (BODIPY)-based photosensitizers for photodynamic therapy, RSC Adv., 2, 11169, 10.1039/c2ra21404k Agostinis, 2011, Photodynamic therapy of cancer: an update, CA Cancer J. Clin., 61, 250, 10.3322/caac.20114 Richter, 1993, Liposomal Delivery of a Photosensitizer, Benzoporphyrin Derivative Monoacid Ring a (Bpd), to Tumor Tissue in a Mouse Tumor Model, Photochem. Photobiol., 57, 1000, 10.1111/j.1751-1097.1993.tb02962.x Tournas, 2009, Combined benzoporphyrin derivative monoacid ring photodynamic therapy and pulsed dye laser for port wine stain birthmarks, Photodiagn. Photodyn. Ther., 6, 195, 10.1016/j.pdpdt.2009.10.002 Spernyak, 2010, Hexylether derivative of pyropheophorbide-a (HPPH) on conjugating with 3gadolinium (III) aminobenzyldiethylenetriaminepentaacetic acid shows potential for in vivo tumor imaging (MR, fluorescence) and photodynamic therapy, Bioconjug. Chem., 21, 828, 10.1021/bc9005317 Clichici, 2010, The dynamics of reactive oxygen species in photodynamic therapy with tetra sulfophenyl-porphyrin, Acta Physiol. Hung., 97, 41, 10.1556/APhysiol.97.2010.1.5 Nanashima, 2012, Photodynamic therapy using talaporfin sodium (Laserphyrin(R)) for bile duct carcinoma: a preliminary clinical trial, Anticancer Res., 32, 4931 Decker, 2013, Pharmacokinetics of temoporfin-loaded liposome formulations: correlation of liposome and temoporfin blood concentration, J. Control. Release, 166, 277, 10.1016/j.jconrel.2013.01.005 Duchi, 2013, Mesenchymal stem cells as delivery vehicle of porphyrin loaded nanoparticles: effective photoinduced in vitro killing of osteosarcoma, J. Control. Release, 168, 225, 10.1016/j.jconrel.2013.03.012 Valle, 2014, Observational, retrospective study of the effectiveness of 5-aminolevulinic acid in malignant glioma surgery in Spain (The VISIONA study), Neurología (English Edition), 29, 131, 10.1016/j.nrleng.2013.05.004 Rapp, 2014, Endoscopic-assisted visualization of 5-aminolevulinic acid–induced fluorescence in malignant glioma surgery: a technical note, World Neurosurg., 82, e277, 10.1016/j.wneu.2013.07.002 Nowak-Stepniowska, 2013, Photodynamic method of cancer diagnosis and therapy--mechanisms and applications, Postepy Biochem., 59, 53 Kwiatkowski, 2018, Photodynamic therapy - mechanisms, photosensitizers and combinations, Biomed. Pharmacother., 106, 1098, 10.1016/j.biopha.2018.07.049 Morton, 2002, The emerging role of 5-ALA-PDT in dermatology: is PDT superior to standard treatments?, J. Dermatol. Treat., 13, s25, 10.1080/095466302317414672 Chilakamarthi, 2017, Photodynamic Therapy: Past, Present and Future, Chem Rec, 17, 775 Mfouo-Tynga, 2021, Features of third generation photosensitizers used in anticancer photodynamic therapy: Review, Photodiagn. Photodyn. Ther., 34, 102091, 10.1016/j.pdpdt.2020.102091 Lucky, 2015, Nanoparticles in photodynamic therapy, Chem. Rev., 115, 1990, 10.1021/cr5004198 Master, 2013, Photodynamic nanomedicine in the treatment of solid tumors: perspectives and challenges, J. Control. Release, 168, 88, 10.1016/j.jconrel.2013.02.020 Allison, 2010, Future of oncologic photodynamic therapy, Future Oncol., 6, 929, 10.2217/fon.10.51 Maeda, 2000, Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review, J. Control. Release, 65, 271, 10.1016/S0168-3659(99)00248-5 Zhang, 2009, Multifunctional core− shell nanoparticles as highly efficient imaging and photosensitizing agents, Langmuir, 25, 10153, 10.1021/la902235d Nagahara, 2013, Antimicrobial photodynamic therapy using a diode laser with a potential new photosensitizer, indocyanine green-loaded nanospheres, may be effective for the clearance of P orphyromonas gingivalis, J. Periodontal Res., 48, 591, 10.1111/jre.12042 Lee, 2013, Ursodeoxycholic acid-conjugated chitosan for photodynamic treatment of HuCC-T1 human cholangiocarcinoma cells, Int. J. Pharm., 454, 74, 10.1016/j.ijpharm.2013.06.035 Zhu, 2015, Upconversion nanoparticle-mediated photodynamic therapy induces THP-1 macrophage apoptosis via ROS bursts and activation of the mitochondrial caspase pathway, Int. J. Nanomedicine, 10, 3719 Jinfeng Zeng, 2018 Aggad, 2018, Gemcitabine delivery and photodynamic therapy in cancer cells via porphyrin-ethylene-based periodic mesoporous organosilica nanoparticles, ChemNanoMat, 4, 46, 10.1002/cnma.201700264 Liang, 2020, Perfluorocarbon@ Porphyrin nanoparticles for tumor hypoxia relief to enhance photodynamic therapy against liver metastasis of colon cancer, ACS Nano, 14, 13569, 10.1021/acsnano.0c05617 Borzęcka, 2021, Encapsulation of glycosylated porphyrins in silica nanoparticles to enhance the efficacy of cancer photodynamic therapy, Mater. Adv., 2, 1613, 10.1039/D0MA00830C Magaela, 2022, Sn (IV) porphyrin-biotin decorated nitrogen doped graphene quantum dots nanohybrids for photodynamic therapy, Polyhedron, 213, 115624, 10.1016/j.poly.2021.115624 Chang, 2021, Porphyrin-lipid stabilized paclitaxel nanoemulsion for combined photodynamic therapy and chemotherapy, J. Nanobiotechnol., 19, 1, 10.1186/s12951-021-00898-1 Borzęcka, 2022, Spherical and rod shaped mesoporous silica nanoparticles for cancer-targeted and photosensitizer delivery in photodynamic therapy, J. Mater. Chem. B, 10, 3248, 10.1039/D1TB02299G Li, 2022, Water-Soluble Porphyrin-Based Nanoparticles Derived from Electrostatic Interaction for Enhanced Photodynamic Therapy, ACS Appl. Bio Mater., 5, 881, 10.1021/acsabm.1c01262 Kurokawa, 2022, Near-Infrared Light Irradiation of Porphyrin-Modified Gold Nanoparticles Promotes Cancer-Cell-Specific Cytotoxicity, Molecules, 27, 1238, 10.3390/molecules27041238 Makola, 2022, Photodynamic therapy activity of 5, 10, 15-tris (5-bromo-2-thienyl), 20 (phenylcarboxy) porphyrin conjugated to graphene quantum dot against MCF-7 breast cancer cells, J. Coord. Chem., 75, 1112, 10.1080/00958972.2022.2087515 Li, 2022, Self-assembled porphyrin polymer nanoparticles with NIR-II emission and highly efficient photothermal performance in cancer therapy, Mater. Today Bio., 13, 100198, 10.1016/j.mtbio.2021.100198 Su, 2022, Self-assembling porphyrin conjugate-carboplatin (IV) prodrug nanoparticles for enhancing high efficacy nasopharyngeal cancer and low systemic toxicity, J. Biomater. Sci. Polym. Ed., 33, 1828, 10.1080/09205063.2022.2087275 Cheng, 2023, Self-assembled porphyrin-based photosensitizer nanomicelles for enhanced photodynamic therapy, Biochem. Biophys. Res. Commun., 652, 55, 10.1016/j.bbrc.2023.02.027 Hameed, 2023, Ultrasmall porphyrin-silica core–shell dots for enhanced fluorescence imaging-guided cancer photodynamic therapy, Nanoscale Adv., 5, 277, 10.1039/D2NA00704E Liang, 2023, Reduction of Platinum (IV) Prodrug Hemoglobin Nanoparticles with Deeply Penetrating Ultrasound Radiation for Tumor-Targeted Therapeutically Enhanced Anticancer Therapy, Angew. Chem. Int. Ed., 62, 10.1002/anie.202301074 Zhao, 2004, Development of organic-dye-doped silica nanoparticles in a reverse microemulsion, Adv. Mater., 16, 173, 10.1002/adma.200305622 Li, 2021, Metal-free nanoassemblies of water-soluble photosensitizer and adenosine triphosphate for efficient and precise photodynamic cancer therapy, ACS Nano, 15, 4979, 10.1021/acsnano.0c09913 Guo, 2022, Tuning the aqueous self-assembly of porphyrins by varying the number of cationic side chains, J. Mater. Chem. B, 10, 5968, 10.1039/D2TB00720G Ni, 2018, Magnetic targeting of nanotheranostics enhances cerenkov radiation-induced photodynamic therapy, J. Am. Chem. Soc., 140, 14971, 10.1021/jacs.8b09374 Daouk, 2020, Can Cerenkov light really induce an effective photodynamic therapy?, Radiation, 1, 5, 10.3390/radiation1010002 Qian, 2022, Minimizing adverse effects of Cerenkov radiation induced photodynamic therapy with transformable photosensitizer-loaded nanovesicles, J. Nanobiotechnol., 20, 203, 10.1186/s12951-022-01401-0 Spinelli, 2021, Photodynamic therapy using Cerenkov and radioluminescence light, Front. Phys., 9, 637120, 10.3389/fphy.2021.637120 Yin, 2022, Engineering bacteria and bionic bacterial derivatives with nanoparticles for cancer therapy, Small, 18, 2104643, 10.1002/smll.202104643 Yan, 2021, Application of phototherapeutic-based nanoparticles in colorectal cancer, Int. J. Biol. Sci., 17, 1361, 10.7150/ijbs.58773 Lin, 2021, Inorganic Nanomaterial-Mediated Gene Therapy in Combination with Other Antitumor Treatment Modalities, Adv. Funct. Mater., 31, 2007096, 10.1002/adfm.202007096 Zhou, 2021, Supramolecular cancer nanotheranostics, Chem. Soc. Rev., 50, 2839, 10.1039/D0CS00011F Nordmann, 2021, 5-Aminolevulinic acid radiodynamic therapy for treatment of high-grade gliomas: A systematic review, Clin. Neurol. Neurosurg., 201, 106430, 10.1016/j.clineuro.2020.106430 Saravanakumar, 2017, Reactive-oxygen-species-responsive drug delivery systems: promises and challenges, Adv. Sci., 4, 1600124, 10.1002/advs.201600124 Fan, 2019, Breaking the depth dependence by nanotechnology-enhanced X-ray-excited deep cancer theranostics, Adv. Mater., 31, 1806381, 10.1002/adma.201806381 Xu, 2022, Recent trends of mesoporous silica-based nanoplatforms for nanodynamic therapies, Coord. Chem. Rev., 469, 214687, 10.1016/j.ccr.2022.214687 Sun, 2022, Nanosensitizer-Mediated Unique Dynamic Therapy Tactics for Effective Inhibition of Deep Tumors, Adv. Drug Deliv. Rev., 114643 Xie, 2021, Application of new radiosensitizer based on nano-biotechnology in the treatment of glioma, Front. Oncol., 11, 633827, 10.3389/fonc.2021.633827 Clement, 2020, Mechanisms for tuning engineered nanomaterials to enhance radiation therapy of cancer, Adv. Sci., 7, 2003584, 10.1002/advs.202003584 Xu, 2016, Combination of photodynamic therapy with radiotherapy for cancer treatment, J. Nanomater., 2016, 10.1155/2016/8507924 Mandl, 2023, Combining Pr3+-Doped Nanoradiosensitizers and Endogenous Protoporphyrin IX for X-ray-Mediated Photodynamic Therapy of Glioblastoma Cells, ACS Appl. Bio Mater., 6, 2370, 10.1021/acsabm.3c00201 Panetta, 2020, Radiodynamic therapy using 15-MV radiation combined with 5-aminolevulinic acid and carbamide peroxide for prostate cancer in vivo, Phys. Med. Biol., 65, 165008, 10.1088/1361-6560/ab9776 Pignatelli, 2023, Reactive Oxygen Species Produced by 5-Aminolevulinic Acid Photodynamic Therapy in the Treatment of Cancer, Int. J. Mol. Sci., 24, 8964, 10.3390/ijms24108964 Gunaydin, 2021, Photodynamic therapy—current limitations and novel approaches, Front. Chem., 9, 691697, 10.3389/fchem.2021.691697 Yang, 2022, Therapeutic effects of in-vivo radiodynamic therapy (RDT) for lung cancer treatment: a combination of 15MV photons and 5-aminolevulinic acid (5-ALA), Biomed. Phys. & Eng. Express, 8, 10.1088/2057-1976/ac9b5c Smith, 2011, Mono-and tri-cationic porphyrin–monoclonal antibody conjugates: photodynamic activity and mechanism of action, Immunology, 132, 256, 10.1111/j.1365-2567.2010.03359.x Obaid, 2015, Cancer targeting with biomolecules: a comparative study of photodynamic therapy efficacy using antibody or lectin conjugated phthalocyanine-PEG gold nanoparticles, Photochem. Photobiol. Sci., 14, 737, 10.1039/c4pp00312h Bryden, 2018, Assembly of high-potency photosensitizer–antibody conjugates through application of dendron multiplier technology, Bioconjug. Chem., 29, 176, 10.1021/acs.bioconjchem.7b00678 Sadraeian, 2021, Photoinduced photosensitizer–antibody conjugates kill HIV env-expressing cells, also inactivating HIV, ACS Omega, 6, 16524, 10.1021/acsomega.1c01721 Kang, 2017, Photothermal therapeutic application of gold nanorods-porphyrin-trastuzumab complexes in HER2-positive breast cancer, Sci. Rep., 7, 42069, 10.1038/srep42069 Penon, 2017, Water soluble, multifunctional antibody-porphyrin gold nanoparticles for targeted photodynamic therapy, J. Colloid Interface Sci., 496, 100, 10.1016/j.jcis.2017.02.006 Narsireddy, 2015, Photosensitizer and peptide-conjugated PAMAM dendrimer for targeted in vivo photodynamic therapy, Int. J. Nanomedicine, 6865 Kamarulzaman, 2015, New peptide-conjugated chlorin-type photosensitizer targeting neuropilin-1 for anti-vascular targeted photodynamic therapy, Int. J. Mol. Sci., 16, 24059, 10.3390/ijms161024059 Li, 2018, cRGD peptide-conjugated pyropheophorbide-a photosensitizers for tumor targeting in photodynamic therapy, Mol. Pharm., 15, 1505, 10.1021/acs.molpharmaceut.7b01064 Yan, 2021, CD133 peptide-conjugated pyropheophorbide-a as a novel photosensitizer for targeted photodynamic therapy in colorectal cancer stem cells, Biomater. Sci., 9, 2020, 10.1039/D0BM01874K Chu, 2020, Facile one-pot synthesis of cyclic peptide-conjugated photosensitisers for targeted photodynamic therapy, Chem. Commun., 56, 11941, 10.1039/D0CC05264G Dos Santos, 2019, Photodynamic therapy in cancer treatment-an update review, J.. Cancer Metastasis Treat, 5 Montaseri, 2020, Organic nanoparticle based active targeting for photodynamic therapy treatment of breast cancer cells, Oncotarget, 11, 2120, 10.18632/oncotarget.27596 Plaetzer, 2005, Apoptosis following photodynamic tumor therapy: induction, mechanisms and detection, Curr. Pharm. Des., 11, 1151, 10.2174/1381612053507648 Robertson, 2009, Photodynamic therapy (PDT): a short review on cellular mechanisms and cancer research applications for PDT, J. Photochem. Photobiol. B Biol., 96, 1, 10.1016/j.jphotobiol.2009.04.001 Agnez-Lima, 2012, DNA damage by singlet oxygen and cellular protective mechanisms, Mutat. Res./Rev. Mutat. Res., 751, 15, 10.1016/j.mrrev.2011.12.005 Ouyang, 2012, Programmed cell death pathways in cancer: a review of apoptosis, autophagy and programmed necrosis, Cell Prolif., 45, 487, 10.1111/j.1365-2184.2012.00845.x Mroz, 2011, Cell death pathways in photodynamic therapy of cancer, Cancers, 3, 2516, 10.3390/cancers3022516 Mishchenko, 2022, Which cell death modality wins the contest for photodynamic therapy of cancer?, Cell Death Dis., 13, 455, 10.1038/s41419-022-04851-4 Dolmans, 2003, Photodynamic therapy for cancer, Nat. Rev. Cancer, 3, 380, 10.1038/nrc1071 Agostinis, 2011, Photodynamic therapy of cancer: an update, CA Cancer J. Clin., 61, 250, 10.3322/caac.20114 Soldani, 2007, Apoptosis as a consequence of multiple organelle damage in cultured tumor cells after photosensitization with rose Bengal acetate, Cell Apopt. and Cancer, 23 Khalil, 2020, Interruption of autophagosome formation in cardiovascular disease, an evidence for protective response of autophagy, Immunol. Investig., 49, 249, 10.1080/08820139.2019.1635619 Bampton, 2005, The dynamics of autophagy visualised in live cells: from autophagosome formation to fusion with endo/lysosomes, Autophagy, 1, 23, 10.4161/auto.1.1.1495 Bordi, 2019, mTOR hyperactivation in Down Syndrome underlies deficits in autophagy induction, autophagosome formation, and mitophagy, Cell Death Dis., 10, 563, 10.1038/s41419-019-1752-5 Tanida, 2011, Autophagosome formation and molecular mechanism of autophagy, Antioxid. Redox Signal., 14, 2201, 10.1089/ars.2010.3482 Sun, 2020, The emerging role of ferroptosis in inflammation, Biomed. Pharmacother., 127, 110108, 10.1016/j.biopha.2020.110108 Luo, 2021, Targeting ferroptosis-based cancer therapy using nanomaterials: strategies and applications, Theranostics, 11, 9937, 10.7150/thno.65480 Zhu, 2022, Ferroptosis-Driven Nanotherapeutics to Reverse Drug Resistance in Tumor Microenvironment, ACS Appl. Bio Mater., 5, 2481, 10.1021/acsabm.2c00199 Tan, 2023, Glutathione system enhancement for cardiac protection: pharmacological options against oxidative stress and ferroptosis, Cell Death Dis., 14, 131, 10.1038/s41419-023-05645-y Xiong, 2021, Engineering nanomedicine for glutathione depletion-augmented cancer therapy, Chem. Soc. Rev., 50, 6013, 10.1039/D0CS00718H Niu, 2021, Application of glutathione depletion in cancer therapy: Enhanced ROS-based therapy, ferroptosis, and chemotherapy, Biomaterials, 277, 121110, 10.1016/j.biomaterials.2021.121110 Kajarabille, 2019, Programmed cell-death by ferroptosis: antioxidants as mitigators, Int. J. Mol. Sci., 20, 4968, 10.3390/ijms20194968 Latunde-Dada, 2017, Ferroptosis: role of lipid peroxidation, iron and ferritinophagy, Biochim. Biophys. Acta (BBA)-General Subj., 1861, 1893, 10.1016/j.bbagen.2017.05.019 Ros, 2020, Partners in crime: the interplay of proteins and membranes in regulated necrosis, Int. J. Mol. Sci., 21, 2412, 10.3390/ijms21072412 Naowarojna, 2023, Dynamic regulation of ferroptosis by lipid metabolism, Antioxid. Redox Signal., 39, 59, 10.1089/ars.2023.0278 Liu, 2023, Ferroptosis: The Entanglement between Traditional Drugs and Nanodrugs in Tumor Therapy, Adv. Healthc. Mater., 12, 2203085, 10.1002/adhm.202203085 Su, 2019, Reactive oxygen species-induced lipid peroxidation in apoptosis, autophagy, and ferroptosis, Oxidative Med. Cell. Longev., 2019, 10.1155/2019/5080843 Chen, 2021, Cellular degradation systems in ferroptosis, Cell Death Differ., 28, 1135, 10.1038/s41418-020-00728-1 Buytaert, 2006, Role of endoplasmic reticulum depletion and multidomain proapoptotic BAX and BAK proteins in shaping cell death after hypericin-mediated photodynamic therapy, FASEB J., 20, 756, 10.1096/fj.05-4305fje Chilakamarthi, 2017, Photodynamic therapy: past, present and future, Chem. Rec., 17, 775, 10.1002/tcr.201600121 Golstein, 2007, Cell death by necrosis: towards a molecular definition, Trends Biochem. Sci., 32, 37, 10.1016/j.tibs.2006.11.001 Richards, 2012, Prognostic value of tumour necrosis and host inflammatory responses in colorectal cancer, J. British Surg., 99, 287, 10.1002/bjs.7755 Panzarini, 2013, Immunogenic cell death: can it be exploited in PhotoDynamic Therapy for cancer?, Biomed. Res. Int., 2013, 10.1155/2013/482160 Turubanova, 2019, Immunogenic cell death induced by a new photodynamic therapy based on photosens and photodithazine, J. Immunotherap. Cancer, 7, 1, 10.1186/s40425-019-0826-3 Ogawa, 2017, Immunogenic cancer cell death selectively induced by near infrared photoimmunotherapy initiates host tumor immunity, Oncotarget, 8, 10425, 10.18632/oncotarget.14425 Alzeibak, 2021, Targeting immunogenic cancer cell death by photodynamic therapy: past, present and future, J. Immunotherap. Cancer, 9, 10.1136/jitc-2020-001926 Kroemer, 2013, Immunogenic cell death in cancer therapy, Annu. Rev. Immunol., 31, 51, 10.1146/annurev-immunol-032712-100008 Wang, 2018, Immunogenic effects of chemotherapy-induced tumor cell death, Genes & Diseases, 5, 194, 10.1016/j.gendis.2018.05.003 Iulianna, 2022, The Achilles’ heel of cancer: targeting tumors via lysosome-induced immunogenic cell death, Cell Death Dis., 13, 509, 10.1038/s41419-022-04912-8 Bianchi, 2017, High-mobility group box 1 protein orchestrates responses to tissue damage via inflammation, innate and adaptive immunity, and tissue repair, Immunol. Rev., 280, 74, 10.1111/imr.12601 Ahmed, 2020, Targeting immunogenic cell death in cancer, Mol. Oncol., 14, 2994, 10.1002/1878-0261.12851 Garg, 2017, Cell death and immunity in cancer: from danger signals to mimicry of pathogen defense responses, Immunol. Rev., 280, 126, 10.1111/imr.12574 Chen, 2022, Nanomaterials: small particles show huge possibilities for cancer immunotherapy, J. Nanobiotechnol., 20, 1, 10.1186/s12951-022-01692-3 Prieto, 2015, Immunological landscape and immunotherapy of hepatocellular carcinoma, Nat. Rev. Gastroenterol. Hepatol., 12, 681, 10.1038/nrgastro.2015.173 Choi, 2021, Mechanism of EBV inducing anti-tumour immunity and its therapeutic use, Nature, 590, 157, 10.1038/s41586-020-03075-w Xu, 2022, Targeted nanomedicines remodeling immunosuppressive tumor microenvironment for enhanced cancer immunotherapy, Acta Pharm. Sin. B, 12, 4327, 10.1016/j.apsb.2022.11.001 Gao, 2021, Nanotechnology for boosting cancer immunotherapy and remodeling tumor microenvironment: the horizons in cancer treatment, ACS Nano, 15, 12567, 10.1021/acsnano.1c02103 Gu, 2022, Photodynamic therapy improves the clinical efficacy of advanced colorectal cancer and recruits immune cells into the tumor immune microenvironment, Front. Immunol., 13, 1050421, 10.3389/fimmu.2022.1050421 Lu, 2023, Immuno-photodynamic Therapy (IPDT): Organic Photosensitizers and Their Application in Cancer Ablation, JACS Au, 3, 682, 10.1021/jacsau.2c00591 Zhang, 2022, How to improve photodynamic therapy-induced antitumor immunity for cancer treatment?, Theranostics, 12, 4629, 10.7150/thno.72465 Xue, 2019, Porphyrin-based nanomedicines for cancer treatment, Bioconjug. Chem., 30, 1585, 10.1021/acs.bioconjchem.9b00231 Liu, 2020, Construction of carrier-free porphyrin-based drug self-framed delivery system to reverse multidrug resistance through photodynamic-chemotherapy, Dyes Pigments, 177, 107922, 10.1016/j.dyepig.2019.107922 Montaseri, 2020, Recent advances in porphyrin-based inorganic nanoparticles for cancer treatment, Int. J. Mol. Sci., 21, 3358, 10.3390/ijms21093358 Li, 2019, Adenosine triphosphate templated self-assembly of cationic porphyrin into chiral double superhelices and enzyme-mediated disassembly, J. Am. Chem. Soc., 141, 12610, 10.1021/jacs.9b04133 Qindeel, 2021, Porphyrin-based nanostructures for cancer theranostics: Chemistry, fundamentals and recent advances, ChemistrySelect, 6, 14082, 10.1002/slct.202103418 Li, 2022, Reaction mechanism of nanomedicine based on porphyrin skeleton and its application prospects, Photodiagn. Photodyn. Ther., 103236 Fan, 2022, Oxygen self-supplied enzyme nanogels for tumor targeting with amplified synergistic starvation and photodynamic therapy, Acta Biomater., 142, 274, 10.1016/j.actbio.2022.01.056 Akhter, 2021, Porphyrin-Based Nanomaterials for Cancer Nanotheranostics, Cancer Nanotheranostics: Volume, 1, 275, 10.1007/978-3-030-74330-7_9 Chen, 2015, Nanomedicine for targeted photothermal cancer therapy: where are we now?, Nanomedicine, 10, 1, 10.2217/nnm.14.186 An, 2022, An unexpected strategy to alleviate hypoxia limitation of photodynamic therapy by biotinylation of photosensitizers, Nat. Commun., 13, 2225, 10.1038/s41467-022-29862-9 Sun, 2022, Boosting Type-I and Type-II ROS Production of Water-Soluble Porphyrin for Efficient Hypoxic Tumor Therapy, Mol. Pharm., 20, 606, 10.1021/acs.molpharmaceut.2c00822 Luo, 2017, “One-pot” fabrication of highly versatile and biocompatible poly (vinyl alcohol)-porphyrin-based nanotheranostics, Theranostics, 7, 3901, 10.7150/thno.20190 Guo, 2016, A porphyrin-based conjugated polymer for highly efficient in vitro and in vivo photothermal therapy, Small, 12, 6243, 10.1002/smll.201602293