DOI QR코드

DOI QR Code

Clinical development of photodynamic agents and therapeutic applications

  • Baskaran, Rengarajan (World Class Smart Lab, Department of New Drug Development, Inha University College of Medicine) ;
  • Lee, Junghan (World Class Smart Lab, Department of New Drug Development, Inha University College of Medicine) ;
  • Yang, Su-Geun (World Class Smart Lab, Department of New Drug Development, Inha University College of Medicine)
  • Received : 2018.07.12
  • Accepted : 2018.09.11
  • Published : 2018.12.31

Abstract

Background: Photodynamic therapy (PDT) is photo-treatment of malignant or benign diseases using photosensitizing agents, light, and oxygen which generates cytotoxic reactive oxygens and induces tumour regressions. Several photodynamic treatments have been extensively studied and the photosensitizers (PS) are key to their biological efficacy, while laser and oxygen allow to appropriate and flexible delivery for treatment of diseases. Introduction: In presence of oxygen and the specific light triggering, PS is activated from its ground state into an excited singlet state, generates reactive oxygen species (ROS) and induces apoptosis of cancer tissues. Those PS can be divided by its specific efficiency of ROS generation, absorption wavelength and chemical structure. Main body: Up to dates, several PS were approved for clinical applications or under clinical trials. $Photofrin^{(R)}$ is the first clinically approved photosensitizer for the treatment of cancer. The second generation of PS, Porfimer sodium ($Photofrin^{(R)}$), Temoporfin ($Foscan^{(R)}$), Motexafin lutetium, Palladium bacteriopheophorbide, $Purlytin^{(R)}$, Verteporfin ($Visudyne{(R)}$), Talaporfin ($Laserphyrin^{(R)}$) are clinically approved or under-clinical trials. Now, third generation of PS, which can dramatically improve cancer-targeting efficiency by chemical modification, nano-delivery system or antibody conjugation, are extensively studied for clinical development. Conclusion: Here, we discuss up-to-date information on FDA-approved photodynamic agents, the clinical benefits of these agents. However, PDT is still dearth for the treatment of diseases in specifically deep tissue cancer. Next generation PS will be addressed in the future for PDT. We also provide clinical unmet need for the design of new photosensitizers.

Keywords

Acknowledgement

Supported by : National Research Foundation of Korea (NRF), WCSL (World Class Smart Lab)

References

  1. Usuda J, Kato H, Okunaka T, Furukawa K, Tsutsui H, Yamada K, et al. Photodynamic therapy (PDT) for lung cancers. J Thorac Oncol. 2006;1:489-93. https://doi.org/10.1016/S1556-0864(15)31616-6
  2. Dolmans DE, Fukumura D, Jain RK. Photodynamic therapy for cancer. Nat Rev Cancer. 2003;3:380-7. https://doi.org/10.1038/nrc1071
  3. Brown SB, Brown EA, Walker I. The present and future role of photodynamic therapy in cancer treatment. Lancet Oncol. 2004;5:497-508. https://doi.org/10.1016/S1470-2045(04)01529-3
  4. Pandey RK, Goswami LN, Chen Y, Gryshuk A, Missert JR, Oseroff A, et al. Nature: a rich source for developing multifunctional agents. Tumor-imaging and photodynamic therapy. Lasers Surg Med. 2006;38:445-67. https://doi.org/10.1002/lsm.20352
  5. Leman JA, Morton CA. Photodynamic therapy: applications in dermatology. Expert Opin Biol Ther. 2002;2:45-53. https://doi.org/10.1517/14712598.2.1.45
  6. Photodynamic therapy with verteporfin for age-related macular degeneration. American Academy of ophthalmology. Ophthalmology. 2000;107:2314-7. https://doi.org/10.1016/S0161-6420(00)00562-5
  7. Rubio N, Rajadurai A, Held KD, Prise KM, Liber HL, Redmond RW. Real-time imaging of novel spatial and temporal responses to photodynamic stress. Free Radic Biol Med. 2009;47:283-90. https://doi.org/10.1016/j.freeradbiomed.2009.04.024
  8. Kolarova H, Nevrelova P, Tomankova K, Kolar P, Bajgar R, Mosinger J. Production of reactive oxygen species after photodynamic therapy by porphyrin sensitizers. Gen Physiol Biophy. 2008;27:101-5.
  9. Daniell MD, Hill JS. A history of photodynamic therapy. Aust N Z J Surg. 1991;61:340-8. https://doi.org/10.1111/j.1445-2197.1991.tb00230.x
  10. Kou J, Dou D, Yang L. Porphyrin photosensitizers in photodynamic therapy and its applications. Oncotarget. 2017;8:81591-603.
  11. Evensen JF. The use of porphyrins and non-ionizing radiation for treatment of cancer. Acta Oncol. 1995;34:1103-10. https://doi.org/10.3109/02841869509127237
  12. Kelly JF, Snell ME. Hematoporphyrin derivative: a possible aid in the diagnosis and therapy of carcinoma of the bladder. J Urol. 1976;115:150-1. https://doi.org/10.1016/S0022-5347(17)59108-9
  13. Hayata Y, Kato H, Konaka C, Ono J, Takizawa N. Hematoporphyrin derivative and laser photoradiation in the treatment of lung cancer. Chest. 1982;81:269-77. https://doi.org/10.1378/chest.81.3.269
  14. Dougherty TJ, Kaufman JE, Goldfarb A, Weishaupt KR, Boyle D, Mittleman A. Photoradiation therapy for the treatment of malignant tumors. Cancer Res. 1978;38:2628-35.
  15. Castano AP, Demidova TN, Hamblin MR. Mechanisms in photodynamic therapy: part one-photosensitizers, photochemistry and cellular localization. Photodiagn Photodyn Ther. 2004;1:279-93. https://doi.org/10.1016/S1572-1000(05)00007-4
  16. Vileno B, Sienkiewicz A, Lekka M, Kulik AJ, Forro L. In vitro assay of singlet oxygen generation in the presence of water-soluble derivatives of C60. Carbon. 2004;42:1195-8. https://doi.org/10.1016/j.carbon.2003.12.042
  17. Foote CS. Mechanisms of photosensitized oxidation. Science. 1968;162:963-70. https://doi.org/10.1126/science.162.3857.963
  18. Vaupel P, Hockel M, Mayer A. Detection and characterization of tumor hypoxia using pO2 histography. Antioxid Redox Signal. 2007;9:1221-35. https://doi.org/10.1089/ars.2007.1628
  19. Nordsmark M, Bentzen SM, Overgaard J. Measurement of human tumour oxygenation status by a polarographic needle electrode. An analysis of inter- and intratumour heterogeneity. Acta Oncol. 1994;33:383-9. https://doi.org/10.3109/02841869409098433
  20. Cui J, Mao X, Olman V, Hastings PJ, Xu Y. Hypoxia and miscoupling between reduced energy efficiency and signaling to cell proliferation drive cancer to grow increasingly faster. J Mol Cell Biol. 2012;4:174-6. https://doi.org/10.1093/jmcb/mjs017
  21. McKeown SR. Defining normoxia, physoxia and hypoxia in tumoursimplications for treatment response. Br J Radiol. 2014;87:20130676. https://doi.org/10.1259/bjr.20130676
  22. Meuthen D, Rick IP, Thunken T, Baldauf SA. Visual prey detection by nearinfrared cues in a fish. Die Naturwissenschaften. 2012;99:1063-6. https://doi.org/10.1007/s00114-012-0980-7
  23. Keereweer S, Van Driel PB, Robinson DJ, Lowik CW. Shifting focus in optical image-guided cancer therapy. Mol Imaging Biol 2014; 16:1-9. https://doi.org/10.1007/s11307-013-0688-x
  24. Rasmussen-Taxdal DS, Ward GE, Figge FH. Fluorescence of human lymphatic and cancer tissues following high doses of intravenous hematoporphyrin. Surg Forum. 1955;5:619-24.
  25. Allison RR, Bagnato VS, Cuenca R, Downie GH, Sibata CH. The future of photodynamic therapy in oncology. Future Oncol. 2006;2:53-71. https://doi.org/10.2217/14796694.2.1.53
  26. Bellnier DA, Greco WR, Loewen GM, Nava H, Oseroff AR, Dougherty TJ. Clinical pharmacokinetics of the PDT photosensitizers porfimer sodium (Photofrin), 2-[1-hexyloxyethyl]-2-devinyl pyropheophorbide-a (Photochlor) and 5-ALA-induced protoporphyrin IX. Lasers Surg Med. 2006;38:439-44. https://doi.org/10.1002/lsm.20340
  27. Reynolds T. Photodynamic therapy expands its horizons. J Natl Cancer Inst. 1997;89:112-4. https://doi.org/10.1093/jnci/89.2.112
  28. Allison RR, Sibata CH. Oncologic photodynamic therapy photosensitizers: a clinical review. Photodiagn Photodyn Ther. 2010;7:61-75. https://doi.org/10.1016/j.pdpdt.2010.02.001
  29. Breskey JD, Lacey SE, Vesper BJ, Paradise WA, Radosevich JA, Colvard MD. Photodynamic therapy: occupational hazards and preventative recommendations for clinical administration by healthcare providers. Photomed Laser Surg. 2013;31:398-407. https://doi.org/10.1089/pho.2013.3496
  30. Schweitzer VG. PHOTOFRIN-mediated photodynamic therapy for treatment of early stage oral cavity and laryngeal malignancies. Lasers Surg Med. 2001;29:305-13. https://doi.org/10.1002/lsm.1133
  31. Lou PJ, Jager HR, Jones L, Theodossy T, Bown SG, Hopper C. Interstitial photodynamic therapy as salvage treatment for recurrent head and neck cancer. Br J Cancer. 2004;91:441-6. https://doi.org/10.1038/sj.bjc.6601993
  32. Story W, Sultan AA, Bottini G, Vaz F, Lee G, Hopper C. Strategies of airway management for head and neck photo-dynamic therapy. Lasers Surg Med. 2013;45:370-6. https://doi.org/10.1002/lsm.22149
  33. Bown SG, Rogowska AZ, Whitelaw DE, Lees WR, Lovat LB, Ripley P, et al. Photodynamic therapy for cancer of the pancreas. Gut. 2002;50:549-57. https://doi.org/10.1136/gut.50.4.549
  34. Pereira SP, Ayaru L, Rogowska A, Mosse A, Hatfield AR, Bown SG. Photodynamic therapy of malignant biliary strictures using mesotetrahydroxyphenylchlorin. Eur J Gastroenterol Hepatol. 2007;19:479-85. https://doi.org/10.1097/MEG.0b013e328013c0bd
  35. Wyss P, Schwarz V, Dobler-Girdziunaite D, Hornung R, Walt H, Degen A, et al. Photodynamic therapy of locoregional breast cancer recurrences using a chlorin-type photosensitizer. Int J Cancer. 2001;93:720-4. https://doi.org/10.1002/ijc.1400
  36. Moore CM, Nathan TR, Lees WR, Mosse CA, Freeman A, Emberton M, et al. Photodynamic therapy using meso tetra hydroxy phenyl chlorin (mTHPC) in early prostate cancer. Lasers Surg Med. 2006;38:356-63. https://doi.org/10.1002/lsm.20275
  37. Nathan TR, Whitelaw DE, Chang SC, Lees WR, Ripley PM, Payne H, et al. Photodynamic therapy for prostate cancer recurrence after radiotherapy: a phase I study. J Urol. 2002;168:1427-32. https://doi.org/10.1016/S0022-5347(05)64466-7
  38. Lorenz KJ, Maier H. Photodynamic therapy with metatetrahydroxyphenylchlorin (Foscan) in the management of squamous cell carcinoma of the head and neck: experience with 35 patients. Eur Arch Otorhinolaryngol. 2009;266:1937-44. https://doi.org/10.1007/s00405-009-0947-2
  39. Patel H, Mick R, Finlay J, Zhu TC, Rickter E, Cengel KA, et al. Motexafin lutetium-photodynamic therapy of prostate cancer: short- and long-term effects on prostate-specific antigen. Clin Cancer Res. 2008;14:4869-76. https://doi.org/10.1158/1078-0432.CCR-08-0317
  40. Finlay JC, Zhu TC, Dimofte A, Stripp D, Malkowicz SB, Whittington R, et al. In vivo determination of the absorption and scattering spectra of the human prostate during photodynamic therapy. Proc SPIE Int Soc Opt Eng. 2014;5315:132-42.
  41. Thomas SR, Khuntia D. Motexafin gadolinium: a promising radiation sensitizer in brain metastasis. Expert Opin Drug Discov. 2011;6:195-203. https://doi.org/10.1517/17460441.2011.546395
  42. Wu GN, Ford JM, Alger JR. MRI measurement of the uptake and retention of motexafin gadolinium in glioblastoma multiforme and uninvolved normal human brain. J Neuro Oncol. 2006;77:95-103. https://doi.org/10.1007/s11060-005-9101-1
  43. Bradley KA, Zhou T, McNall-Knapp RY, Jakacki RI, Levy AS, Vezina G, et al. Motexafin-gadolinium and involved field radiation therapy for intrinsic pontine glioma of childhood: a children's oncology group phase 2 study. Int J Radiat Oncol Biol Phys. 2013;85:e55-60. https://doi.org/10.1016/j.ijrobp.2012.09.004
  44. Bradley KA, Pollack IF, Reid JM, Adamson PC, Ames MM, Vezina G, et al. Motexafin gadolinium and involved field radiation therapy for intrinsic pontine glioma of childhood: a Children's oncology group phase I study. Neuro-Oncology. 2008;10:752-8. https://doi.org/10.1215/15228517-2008-043
  45. Azzouzi AR, Lebdai S, Benzaghou F, Stief C. Vascular-targeted photodynamic therapy with TOOKAD(R) soluble in localized prostate cancer: standardization of the procedure. World J Urol. 2015;33:937-44. https://doi.org/10.1007/s00345-015-1535-2
  46. Eymerit-Morin C, Zidane M, Lebdai S, Triau S, Azzouzi AR, Rousselet MC. Histopathology of prostate tissue after vascular-targeted photodynamic therapy for localized prostate cancer. Virchows Arch. 2013;463:547-52. https://doi.org/10.1007/s00428-013-1454-9
  47. Betrouni N, Boukris S, Benzaghou F. Vascular targeted photodynamic therapy with TOOKAD(R) soluble (WST11) in localized prostate cancer: efficiency of automatic pre-treatment planning. Lasers Med Sci. 2017;32:1301-7. https://doi.org/10.1007/s10103-017-2241-7
  48. Trachtenberg J, Bogaards A, Weersink RA, Haider MA, Evans A, McCluskey SA, et al. Vascular targeted photodynamic therapy with palladiumbacteriopheophorbide photosensitizer for recurrent prostate cancer following definitive radiation therapy: assessment of safety and treatment response. J Urol. 2007;178:1974-9. https://doi.org/10.1016/j.juro.2007.07.036
  49. Taneja SS, Bennett J, Coleman J, Grubb R, Andriole G, Reiter RE, et al. 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. 2016;196:1096-104. https://doi.org/10.1016/j.juro.2016.05.113
  50. Kawczyk-Krupka A, Wawrzyniec K, Musiol SK, Potempa M, Bugaj AM, Sieron A. Treatment of localized prostate cancer using WST-09 and WST-11 mediated vascular targeted photodynamic therapy-a review. Photodiagn Photodyn Ther. 2015;12:567-74. https://doi.org/10.1016/j.pdpdt.2015.10.001
  51. Azzouzi AR, Vincendeau S, Barret E, Cicco A, Kleinclauss F, van der Poel HG, et al. Padeliporfin vascular-targeted photodynamic therapy versus active surveillance in men with low-risk prostate cancer (CLIN1001 PCM301): an open-label, phase 3, randomised controlled trial. Lancet Oncol. 2017;18:181-91. https://doi.org/10.1016/S1470-2045(16)30661-1
  52. Brun PH, DeGroot JL, Dickson EF, Farahani M, Pottier RH. Determination of the in vivo pharmacokinetics of palladium-bacteriopheophorbide (WST09) in EMT6 tumour-bearing Balb/c mice using graphite furnace atomic absorption spectroscopy. Photochem Photobiol Sci. 2004;3:1006-10. https://doi.org/10.1039/b403534h
  53. Weersink RA, Forbes J, Bisland S, Trachtenberg J, Elhilali M, Brun PH, et al. Assessment of cutaneous photosensitivity of TOOKAD (WST09) in preclinical animal models and in patients. Photochem Photobiol. 2005;81:106-13. https://doi.org/10.1562/2004-05-31-RA-182.1
  54. Kaplan MJ, Somers RG, Greenberg RH, Ackler J. Photodynamic therapy in the management of metastatic cutaneous adenocarcinomas: case reports from phase 1/2 studies using tin ethyl etiopurpurin (SnET2). J Surg Oncol. 1998;67:121-5. https://doi.org/10.1002/(SICI)1096-9098(199802)67:2<121::AID-JSO9>3.0.CO;2-C
  55. Mang TS, Allison R, Hewson G, Snider W, Moskowitz R. A phase II/III clinical study of tin ethyl etiopurpurin (Purlytin)-induced photodynamic therapy for the treatment of recurrent cutaneous metastatic breast cancer. Cancer J Sci Am. 1998;4:378-84.
  56. Tsuchihashi T, Mori K, Ueyama K, Yoneya S. Five-year results of photodynamic therapy with verteporfin for Japanese patients with neovascular age-related macular degeneration. Clin Ophthalmol. 2013;7:615-20.
  57. Pece A, Milani P, Isola V, Pierro L. A long-term study of photodynamic therapy with verteporfin for choroidal neovascularization at the edge of chorioretinal atrophy in pathologic myopia. Ophthalmologica. 2011;225:161-8. https://doi.org/10.1159/000322362
  58. Huggett MT, Jermyn M, Gillams A, Illing R, Mosse S, Novelli M, et al. Phase I/II study of verteporfin photodynamic therapy in locally advanced pancreatic cancer. Br J Cancer. 2014;110:1698-704. https://doi.org/10.1038/bjc.2014.95
  59. Yoshida T, Tokashiki R, Ito H, Shimizu A, Nakamura K, Hiramatsu H, et al. Therapeutic effects of a new photosensitizer for photodynamic therapy of early head and neck cancer in relation to tissue concentration. Auris Nasus Larynx. 2008;35:545-51. https://doi.org/10.1016/j.anl.2007.10.008
  60. Shimizu K, Nitta M, Komori T, Maruyama T, Yasuda T, Fujii Y, et al. Intraoperative photodynamic diagnosis using Talaporfin sodium simultaneously applied for photodynamic therapy against malignant glioma: a prospective clinical study. Front Neurol. 2018;9:24. https://doi.org/10.3389/fneur.2018.00024
  61. Hudson R, Carcenac M, Smith K, Madden L, Clarke OJ, Pelegrin A, et al. The development and characterisation of porphyrin isothiocyanate-monoclonal antibody conjugates for photoimmunotherapy. Br J Cancer. 2005;92:1442-9. https://doi.org/10.1038/sj.bjc.6602517
  62. Narumi A, Tsuji T, Shinohara K, Yamazaki H, Kikuchi M, Kawaguchi S, et al. Maltotriose-conjugation to a fluorinated chlorin derivative generating a PDT photosensitizer with improved water-solubility. Org Biomol Chem. 2016;14:3608-13. https://doi.org/10.1039/C6OB00276E
  63. Nishie H, Kataoka H, Yano S, Kikuchi JI, Hayashi N, Narumi A, et al. A nextgeneration bifunctional photosensitizer with improved water-solubility for photodynamic therapy and diagnosis. Oncotarget. 2016;7:74259-68.
  64. Staneloudi C, Smith KA, Hudson R, Malatesti N, Savoie H, Boyle RW, et al. Development and characterization of novel photosensitizer : scFv conjugates for use in photodynamic therapy of cancer. Immunology. 2007;120:512-7. https://doi.org/10.1111/j.1365-2567.2006.02522.x
  65. O'Connor AE, Gallagher WM, Byrne AT. Porphyrin and nonporphyrin photosensitizers in oncology: preclinical and clinical advances in photodynamic therapy. Photochem Photobiol. 2009;85:1053-74. https://doi.org/10.1111/j.1751-1097.2009.00585.x
  66. Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, et al. Photodynamic therapy of cancer: an update. CA Cancer J Clin. 2011;61:250-81. https://doi.org/10.3322/caac.20114
  67. Proll S, Wilhelm B, Robert B, Scheer H. Myoglobin with modified tetrapyrrole chromophores: binding specificity and photochemistry. Biochim Biophys Acta. 2006;1757:750-63. https://doi.org/10.1016/j.bbabio.2006.03.026

Cited by

  1. 808 nm Near-Infrared Light-Excited UCNPs@mSiO 2 -Ce6-GPC3 Nanocomposites For Photodynamic Therapy In Liver Cancer vol.14, pp.None, 2019, https://doi.org/10.2147/ijn.s221496
  2. Multimodal use of the porphyrin TMPyP: From cancer therapy to antimicrobial applications vol.23, pp.1, 2018, https://doi.org/10.1142/s1088424619500111
  3. Characterization of Perturbing Actions by Verteporfin, a Benzoporphyrin Photosensitizer, on Membrane Ionic Currents vol.7, pp.None, 2019, https://doi.org/10.3389/fchem.2019.00566
  4. Mitotic Catastrophe Induced in HeLa Tumor Cells by Photodynamic Therapy with Methyl-aminolevulinate vol.20, pp.5, 2018, https://doi.org/10.3390/ijms20051229
  5. Recent Progress in the Theranostics Application of Nanomedicine in Lung Cancer vol.11, pp.5, 2019, https://doi.org/10.3390/cancers11050597
  6. A Basic Study of Photodynamic Therapy with Glucose-Conjugated Chlorin e6 Using Mammary Carcinoma Xenografts vol.11, pp.5, 2019, https://doi.org/10.3390/cancers11050636
  7. Chlorin e6-Loaded PEG-PCL Nanoemulsion for Photodynamic Therapy and In Vivo Drug Delivery vol.20, pp.16, 2018, https://doi.org/10.3390/ijms20163958
  8. The Role of Photochemical Reactions in the Development of Advanced Soft Materials for Biomedical Applications vol.7, pp.16, 2018, https://doi.org/10.1002/adom.201900215
  9. Hyaluronic Acid-Conjugated with Hyperbranched Chlorin e6 Using Disulfide Linkage and Its Nanophotosensitizer for Enhanced Photodynamic Therapy of Cancer Cells vol.12, pp.19, 2018, https://doi.org/10.3390/ma12193080
  10. Photodynamic activity of Sn(IV) meso -tetraacenaphthylporphyrin and its methyl- $ \beta $ -cyclodextrin inclusion complexes on MCF-7 breast cancer cells vol.23, pp.11, 2019, https://doi.org/10.1142/s1088424619501633
  11. Verteporfin-Loaded Lipid Nanoparticles Improve Ovarian Cancer Photodynamic Therapy In Vitro and In Vivo vol.11, pp.11, 2019, https://doi.org/10.3390/cancers11111760
  12. Glycoconjugated Phthalocyanines as Photosensitizers for PDT – Overcoming Aggregation in Solution vol.2019, pp.42, 2019, https://doi.org/10.1002/ejoc.201901224
  13. Water-Soluble, Zwitterionic Poly-photosensitizers as Carrier-Free, Photosensitizer-Self-Delivery System for in Vivo Photodynamic Therapy vol.11, pp.47, 2018, https://doi.org/10.1021/acsami.9b19546
  14. Photophysicochemical Properties and In Vitro Phototherapeutic Effects of Iodoquinoline- and Benzothiazole-Derived Unsymmetrical Squaraine Cyanine Dyes vol.9, pp.24, 2019, https://doi.org/10.3390/app9245414
  15. Remodeling Tumor Microenvironment by Multifunctional Nanoassemblies for Enhanced Photodynamic Cancer Therapy vol.2, pp.None, 2018, https://doi.org/10.1021/acsmaterialslett.0c00259
  16. Ruthenium Complexes as Anticancer Agents: A Brief History and Perspectives vol.14, pp.None, 2018, https://doi.org/10.2147/dddt.s275007
  17. Multifunctional phototheranostic nanomedicine for cancer imaging and treatment vol.5, pp.None, 2018, https://doi.org/10.1016/j.mtbio.2019.100035
  18. Clinical application of photodynamic therapy for malignant airway tumors in China vol.11, pp.1, 2018, https://doi.org/10.1111/1759-7714.13223
  19. Possible Enhancement of Photodynamic Therapy (PDT) Colorectal Cancer Treatment when Combined with Cannabidiol vol.20, pp.None, 2018, https://doi.org/10.2174/1871520620666200415102321
  20. Efficient photosensitizers with aggregation-induced emission characteristics for lysosome- and Gram-positive bacteria-targeted photodynamic therapy vol.56, pp.17, 2018, https://doi.org/10.1039/d0cc00394h
  21. Nanomolecular singlet oxygen photosensitizers based on hemiquinonoid-resorcinarenes, the fuchsonarenes vol.11, pp.10, 2020, https://doi.org/10.1039/d0sc00651c
  22. Dual-Targeted Phototherapeutic Agents as Magic Bullets for Cancer vol.31, pp.3, 2018, https://doi.org/10.1021/acs.bioconjchem.9b00836
  23. A promising anticancer drug: a photosensitizer based on the porphyrin skeleton vol.11, pp.4, 2020, https://doi.org/10.1039/c9md00558g
  24. EGFR-Targeted Nanobody Functionalized Polymeric Micelles Loaded with mTHPC for Selective Photodynamic Therapy vol.17, pp.4, 2020, https://doi.org/10.1021/acs.molpharmaceut.9b01280
  25. Conjugating biomaterials with photosensitizers: advances and perspectives for photodynamic antimicrobial chemotherapy vol.19, pp.4, 2020, https://doi.org/10.1039/c9pp00398c
  26. Fluorophore-Induced Plasmonic Current: Generation-Based Detection of Singlet Oxygen vol.5, pp.4, 2018, https://doi.org/10.1021/acssensors.0c00377
  27. In vitro photodynamic treatment of cancer cells induced by aza-BODIPYs vol.19, pp.6, 2018, https://doi.org/10.1039/d0pp00026d
  28. Enhanced photodynamic therapy and fluorescence imaging using gold nanorods for porphyrin delivery in a novel in vitro squamous cell carcinoma 3D model vol.8, pp.23, 2020, https://doi.org/10.1039/d0tb00810a
  29. NIR‐II Light Activated Photosensitizer with Aggregation‐Induced Emission for Precise and Efficient Two‐Photon Photodynamic Cancer Cell Ablation vol.30, pp.30, 2018, https://doi.org/10.1002/adfm.202002546
  30. Biodegradable Silica‐Based Nanoparticles with Improved and Safe Delivery of Protoporphyrin IX for the In Vivo Photodynamic Therapy of Breast Cancer vol.3, pp.7, 2018, https://doi.org/10.1002/adtp.202000022
  31. Nanotechnology in Modern Photodynamic Therapy of Cancer: A Review of Cellular Resistance Patterns Affecting the Therapeutic Response vol.12, pp.7, 2018, https://doi.org/10.3390/pharmaceutics12070632
  32. Efficient near-infrared photosensitizer with aggregation-induced emission characteristics for mitochondria-targeted and image-guided photodynamic cancer therapy vol.4, pp.7, 2020, https://doi.org/10.1039/d0qm00170h
  33. NIR Photodynamic Destruction of PDAC and HNSCC Nodules Using Triple-Receptor-Targeted Photoimmuno-Nanoconjugates: Targeting Heterogeneity in Cancer vol.9, pp.8, 2020, https://doi.org/10.3390/jcm9082390
  34. Photodynamic Therapy Using a Novel Phosphorus Tetraphenylporphyrin Induces an Anticancer Effect via Bax/Bcl-xL-related Mitochondrial Apoptosis in Biliary Cancer Cells vol.53, pp.4, 2018, https://doi.org/10.1267/ahc.20-00002
  35. A comparative study of the photophysicochemical and photodynamic activity properties of meso -4-methylthiophenyl functionalized Sn(IV) tetraarylporphyrins and triarylcorroles vol.24, pp.9, 2018, https://doi.org/10.1142/s1088424620500273
  36. A comparative study of the photophysicochemical and photodynamic activity properties of meso -4-methylthiophenyl functionalized Sn(IV) tetraarylporphyrins and triarylcorroles vol.24, pp.9, 2018, https://doi.org/10.1142/s1088424620500273
  37. Antibody-Based Immunotherapy: Alternative Approaches for the Treatment of Metastatic Melanoma vol.8, pp.9, 2020, https://doi.org/10.3390/biomedicines8090327
  38. Role of Photoactive Phytocompounds in Photodynamic Therapy of Cancer vol.25, pp.18, 2018, https://doi.org/10.3390/molecules25184102
  39. Photocytotoxicity of Oligothienyl‐Functionalized Chelates That Sensitize LnIII Luminescence and Generate 1O2 vol.26, pp.52, 2020, https://doi.org/10.1002/chem.202001568
  40. Synthesis and In Vitro Studies of a Gd(DOTA)-Porphyrin Conjugate for Combined MRI and Photodynamic Treatment vol.59, pp.19, 2018, https://doi.org/10.1021/acs.inorgchem.0c02189
  41. Strengthened Tumor Photodynamic Therapy Based on a Visible Nanoscale Covalent Organic Polymer Engineered by Microwave Assisted Synthesis vol.30, pp.45, 2018, https://doi.org/10.1002/adfm.202004834
  42. Photodynamic therapy, priming and optical imaging: Potential co-conspirators in treatment design and optimization - a Thomas Dougherty Award for Excellence in PDT paper vol.24, pp.2, 2020, https://doi.org/10.1142/s1088424620300098
  43. Non-Invasive Diagnostic System Based on Light for Detecting Early-Stage Oral Cancer and High-Risk Precancerous Lesions—Potential for Dentistry vol.12, pp.11, 2020, https://doi.org/10.3390/cancers12113185
  44. Clinical development and potential of photothermal and photodynamic therapies for cancer vol.17, pp.11, 2018, https://doi.org/10.1038/s41571-020-0410-2
  45. Current Targets and Bioconjugation Strategies in Photodynamic Diagnosis and Therapy of Cancer vol.25, pp.21, 2020, https://doi.org/10.3390/molecules25214964
  46. Daylight Photodynamic Therapy: An Update vol.25, pp.21, 2018, https://doi.org/10.3390/molecules25215195
  47. Photodynamic diagnosis and photodynamic therapy of colorectal cancer in vitro and in vivo vol.10, pp.68, 2018, https://doi.org/10.1039/d0ra08617g
  48. Polyacrylic Acid with Methylene Blue Dye as a Sensitizing Agent for Photodynamic Therapy to Reduce Streptococcus mutans in Dentinal Caries vol.38, pp.11, 2018, https://doi.org/10.1089/photob.2019.4736
  49. Pathological and Pharmacological Roles of Mitochondrial Reactive Oxygen Species in Malignant Neoplasms: Therapies Involving Chemical Compounds, Natural Products, and Photosensitizers vol.25, pp.22, 2018, https://doi.org/10.3390/molecules25225252
  50. Heat shock protein 90-targeted photodynamic therapy enables treatment of subcutaneous and visceral tumors vol.3, pp.1, 2018, https://doi.org/10.1038/s42003-020-0956-7
  51. Conjugated Photosensitizers for Imaging and PDT in Cancer Research vol.63, pp.23, 2018, https://doi.org/10.1021/acs.jmedchem.0c00047
  52. Azulitox-A Pseudomonas aeruginosa P28-Derived Cancer-Cell-Specific Protein Photosensitizer vol.21, pp.12, 2018, https://doi.org/10.1021/acs.biomac.0c01216
  53. An update in clinical utilization of photodynamic therapy for lung cancer vol.12, pp.4, 2018, https://doi.org/10.7150/jca.51537
  54. Stimuli‐Responsive Biomaterials: Scaffolds for Stem Cell Control vol.10, pp.1, 2021, https://doi.org/10.1002/adhm.202001125
  55. Recent progress in nanophotosensitizers for advanced photodynamic therapy of cancer vol.4, pp.1, 2021, https://doi.org/10.1088/2515-7639/abc9ce
  56. Photodynamic Therapy for the Treatment and Diagnosis of Cancer-A Review of the Current Clinical Status vol.9, pp.None, 2018, https://doi.org/10.3389/fchem.2021.686303
  57. Remotely Activated Nanoparticles for Anticancer Therapy vol.13, pp.1, 2018, https://doi.org/10.1007/s40820-020-00537-8
  58. Inorganic Nanomaterials for Photothermal‐Based Cancer Theranostics vol.4, pp.2, 2018, https://doi.org/10.1002/adtp.202000207
  59. Near Infrared Photo‐Antimicrobial Targeting Therapy for Candida albicans vol.4, pp.2, 2018, https://doi.org/10.1002/adtp.202000221
  60. Photodynamic therapy exploiting the anti-tumor activity of mannose-conjugated chlorin e6 reduced M2-like tumor-associated macrophages vol.14, pp.2, 2018, https://doi.org/10.1016/j.tranon.2020.101005
  61. Non-Oncologic Applications of Nanomedicine-Based Phototherapy vol.9, pp.2, 2021, https://doi.org/10.3390/biomedicines9020113
  62. Photodynamic Therapy Using Cerenkov and Radioluminescence Light vol.9, pp.None, 2021, https://doi.org/10.3389/fphy.2021.637120
  63. Glycyrrhetinic Acid-Modified Silicon Phthalocyanine for Liver Cancer-Targeted Photodynamic Therapy vol.22, pp.2, 2021, https://doi.org/10.1021/acs.biomac.0c01550
  64. Exploiting a Neutral BODIPY Copolymer as an Effective Agent for Photodynamic Antimicrobial Inactivation vol.125, pp.6, 2018, https://doi.org/10.1021/acs.jpcb.0c09634
  65. Strategies for Cancer Treatment Based on Photonic Nanomedicine vol.14, pp.6, 2018, https://doi.org/10.3390/ma14061435
  66. ICG/L-Arginine Encapsulated PLGA Nanoparticle-Thermosensitive Hydrogel Hybrid Delivery System for Cascade Cancer Photodynamic-NO Therapy with Promoted Collagen Depletion in Tumor Tissue vol.18, pp.3, 2018, https://doi.org/10.1021/acs.molpharmaceut.0c00937
  67. A Review of Bacteriochlorophyllides: Chemical Structures and Applications vol.26, pp.5, 2018, https://doi.org/10.3390/molecules26051293
  68. Phthalocyanine-Conjugated Glyconanoparticles for Chemo-photodynamic Combination Therapy vol.22, pp.4, 2018, https://doi.org/10.1021/acs.biomac.0c01811
  69. Magnetism, Ultrasound, and Light-Stimulated Mesoporous Silica Nanocarriers for Theranostics and Beyond vol.143, pp.16, 2021, https://doi.org/10.1021/jacs.0c10098
  70. Interrogating biological systems using visible-light-powered catalysis vol.5, pp.5, 2021, https://doi.org/10.1038/s41570-021-00265-6
  71. Effect of Cerenkov Radiation-Induced Photodynamic Therapy with 18F-FDG in an Intraperitoneal Xenograft Mouse Model of Ovarian Cancer vol.22, pp.9, 2018, https://doi.org/10.3390/ijms22094934
  72. Photostable Platinated Bacteriochlorins as Potent Photodynamic Agents vol.64, pp.10, 2021, https://doi.org/10.1021/acs.jmedchem.1c00052
  73. Features of third generation photosensitizers used in anticancer photodynamic therapy: Review vol.34, pp.None, 2021, https://doi.org/10.1016/j.pdpdt.2020.102091
  74. Translocator protein (TSPO)-Targeted agents for photodynamic therapy of cancer vol.34, pp.None, 2021, https://doi.org/10.1016/j.pdpdt.2021.102209
  75. Photo-Triggered Nanomaterials for Cancer Theranostic Applications vol.11, pp.2, 2021, https://doi.org/10.1142/s1793984421300041
  76. New Insights into the Clinical Implications of Yes-Associated Protein in Lung Cancer: Roles in Drug Resistance, Tumor Immunity, Autophagy, and Organoid Development vol.13, pp.12, 2018, https://doi.org/10.3390/cancers13123069
  77. Potential of Cyanine Derived Dyes in Photodynamic Therapy vol.13, pp.6, 2021, https://doi.org/10.3390/pharmaceutics13060818
  78. Supramolecular agents for combination of photodynamic therapy and other treatments vol.12, pp.21, 2018, https://doi.org/10.1039/d1sc01125a
  79. Photocytotoxicity of Thiophene- and Bithiophene-Dipicolinato Luminescent Lanthanide Complexes vol.64, pp.11, 2021, https://doi.org/10.1021/acs.jmedchem.0c01805
  80. Comparison of the Photodynamic Action of Porphyrin, Chlorin, and Isobacteriochlorin Derivatives toward a Melanotic Cell Line vol.4, pp.6, 2021, https://doi.org/10.1021/acsabm.1c00218
  81. Insights on the polypyrrole based nanoformulations for photodynamic therapy vol.25, pp.7, 2021, https://doi.org/10.1142/s1088424621300032
  82. Nanophotosensitizers for cancer therapy: a promising technology? vol.4, pp.3, 2018, https://doi.org/10.1088/2515-7639/abf7dd
  83. Plasmonic enhancement of nitric oxide generation vol.13, pp.28, 2018, https://doi.org/10.1039/d1nr02126e
  84. Micro- and Nano-Based Transdermal Delivery Systems of Photosensitizing Drugs for the Treatment of Cutaneous Malignancies vol.14, pp.8, 2018, https://doi.org/10.3390/ph14080772
  85. Conjugated Polymers with Aggregation‐Induced Emission Characteristics for Fluorescence Imaging and Photodynamic Therapy vol.16, pp.15, 2018, https://doi.org/10.1002/cmdc.202100138
  86. Photodynamic therapy using self-assembled nanogels comprising chlorin e6-bearing pullulan vol.9, pp.32, 2021, https://doi.org/10.1039/d1tb00377a
  87. Biodegradable Polymeric Nanoparticles Containing an Immune Checkpoint Inhibitor (aPDL1) to Locally Induce Immune Responses in the Central Nervous System vol.31, pp.38, 2018, https://doi.org/10.1002/adfm.202102274
  88. Pyrrole-based photosensitizers for photodynamic therapy - a Thomas Dougherty award paper vol.25, pp.9, 2018, https://doi.org/10.1142/s1088424621300044
  89. Photodynamic Therapy: A Compendium of Latest Reviews vol.13, pp.17, 2018, https://doi.org/10.3390/cancers13174447
  90. Mito‐Bomb: Targeting Mitochondria for Cancer Therapy vol.33, pp.43, 2021, https://doi.org/10.1002/adma.202007778
  91. Zinc-Phthalocyanine-Loaded Extracellular Vesicles Increase Efficacy and Selectivity of Photodynamic Therapy in Co-Culture and Preclinical Models of Colon Cancer vol.13, pp.10, 2018, https://doi.org/10.3390/pharmaceutics13101547
  92. Advancement of metal compounds as therapeutic and diagnostic metallodrugs: Current frontiers and future perspectives vol.445, pp.None, 2018, https://doi.org/10.1016/j.ccr.2021.214104
  93. Antimicrobial Photodynamic Inactivation Using Topical and Superhydrophobic Sensitizer Techniques: A Perspective from Diffusion in Biofilms vol.97, pp.6, 2018, https://doi.org/10.1111/php.13461
  94. Current Status of Photodynamic Diagnosis for Gastric Tumors vol.11, pp.11, 2018, https://doi.org/10.3390/diagnostics11111967
  95. The Importance of 6-Aminohexanoic Acid as a Hydrophobic, Flexible Structural Element vol.22, pp.22, 2018, https://doi.org/10.3390/ijms222212122
  96. Nanoparticle-Mediated Delivery Systems in Photodynamic Therapy of Colorectal Cancer vol.22, pp.22, 2018, https://doi.org/10.3390/ijms222212405
  97. Nanoformulations-based advancement in the delivery of phytopharmaceuticals for skin cancer management vol.66, pp.None, 2018, https://doi.org/10.1016/j.jddst.2021.102912
  98. Recent Advances in Photodynamic Therapy against Fungal Keratitis vol.13, pp.12, 2021, https://doi.org/10.3390/pharmaceutics13122011
  99. Silver chalcogenide nanoparticles: a review of their biomedical applications vol.13, pp.46, 2021, https://doi.org/10.1039/d0nr03872e
  100. Targeting Cancer Cell Tight Junctions Enhances PLGA-Based Photothermal Sensitizers’ Performance In Vitro and In Vivo vol.14, pp.1, 2018, https://doi.org/10.3390/pharmaceutics14010043
  101. Klinische Entwicklung von Metallkomplexen als Photosensibilisatoren für die photodynamische Therapie von Krebs vol.134, pp.5, 2022, https://doi.org/10.1002/ange.202112236
  102. Clinical Development of Metal Complexes as Photosensitizers for Photodynamic Therapy of Cancer vol.61, pp.5, 2018, https://doi.org/10.1002/anie.202112236