Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Emerging Therapeutic Targets in Oncologic Photodynamic Therapy

Author(s): Gina Manda*, Mihail E. Hinescu, Ionela V. Neagoe, Luis F.V. Ferreira, Rica Boscencu, Paul Vasos, Selma H. Basaga and Antonio Cuadrado

Volume 24, Issue 44, 2018

Page: [5268 - 5295] Pages: 28

DOI: 10.2174/1381612825666190122163832

open access plus

Abstract

Background: Reactive oxygen species sustain tumorigenesis and cancer progression through deregulated redox signalling which also sensitizes cancer cells to therapy. Photodynamic therapy (PDT) is a promising anti-cancer therapy based on a provoked singlet oxygen burst, exhibiting a better toxicological profile than chemo- and radiotherapy. Important gaps in the knowledge on underlining molecular mechanisms impede on its translation towards clinical applications.

Aims and Methods: The main objective of this review is to critically analyse the knowledge lately gained on therapeutic targets related to redox and inflammatory networks underlining PDT and its outcome in terms of cell death and resistance to therapy. Emerging therapeutic targets and pharmaceutical tools will be documented based on the identified molecular background of PDT.

Results: Cellular responses and molecular networks in cancer cells exposed to the PDT-triggered singlet oxygen burst and the associated stresses are analysed using a systems medicine approach, addressing both cell death and repair mechanisms. In the context of immunogenic cell death, therapeutic tools for boosting anti-tumor immunity will be outlined. Finally, the transcription factor NRF2, which is a major coordinator of cytoprotective responses, is presented as a promising pharmacologic target for developing co-therapies designed to increase PDT efficacy.

Conclusion: There is an urgent need to perform in-depth molecular investigations in the field of PDT and to correlate them with clinical data through a systems medicine approach for highlighting the complex biological signature of PDT. This will definitely guide translation of PDT to clinic and the development of new therapeutic strategies aimed at improving PDT.

Keywords: Cancer, photodynamic therapy, reactive oxygen species, oxidative stress, redox signalling, transcription factor NRF2, inflammation.

[1]
Egea J, Fabregat I, Frapart YM, et al. European contribution to the study of ROS: A summary of the findings and prospects for the future from the COST action BM1203 (EU-ROS). Redox Biol 2017; 13: 94-162.
[2]
Cuadrado A, Manda G, Hassan A, et al. Transcription Factor NRF2 as a Therapeutic Target for Chronic Diseases: A Systems Medicine Approach. Pharmacol Rev 2018; 70(2): 348-83.
[3]
Leone A, Roca MS, Ciardiello C, Costantini S, Budillon A. Oxidative Stress Gene Expression Profile Correlates with Cancer Patient Poor Prognosis: Identification of Crucial Pathways Might Select Novel Therapeutic Approaches. Oxid Med Cell Longev 2017; 2017: 2597581.
[4]
Ogrunc M, Di Micco R, Liontos M, et al. Oncogene-induced reactive oxygen species fuel hyperproliferation and DNA damage response activation. Cell Death Differ 2014; 21(6): 998-1012.
[5]
Liou GY, Storz P. Reactive oxygen species in cancer. Free Radic Res 2010; 44(5): 479-96.
[6]
Fiaschi T, Chiarugi P. Oxidative stress, tumor microenvironment, and metabolic reprogramming: A diabolic liaison. Int J Cell Biol 2012; 2012: 762825.
[7]
Burns JS, Manda G. Metabolic Pathways of the Warburg Effect in Health and Disease: Perspectives of Choice, Chain or Chance. Int J Mol Sci 2017; 18(12): E2755.
[8]
Lee M, Yoon JH. Metabolic interplay between glycolysis and mitochondrial oxidation: The reverse Warburg effect and its therapeutic implication. World J Biol Chem 2015; 6(3): 148-61.
[9]
Manda G, Isvoranu G, Comanescu MV, Manea A, Debelec Butuner B, Korkmaz KS. The redox biology network in cancer pathophysiology and therapeutics. Redox Biol 2015; 5: 347-57.
[10]
Redza-Dutordoir M, Averill-Bates DA. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim Biophys Acta 2016; 1863(12): 2977-92.
[11]
Panieri E, Gogvadze V, Norberg E, Venkatesh R, Orrenius S, Zhivotovsky B. Reactive oxygen species generated in different compartments induce cell death, survival, or senescence. Free Radic Biol Med 2013; 57: 176-87.
[12]
Schieber M, Chandel NS. ROS function in redox signaling and oxidative stress. Curr Biol 2014; 24(10): R453-62.
[13]
Sies H. Oxidative stress: A concept in redox biology and medicine. Redox Biol 2015; 4: 180-3.
[14]
Sosa V, Molin A(c)T, Somoza R, Paciucci R, Kondoh H. LLeonart ME. Oxidative stress and cancer: An overview. Ageing Res Rev 2013; 12(1): 376-90.
[15]
Azzam EI, Jay-Gerin JP, Pain D. Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett 2012; 327(1-2): 48-60.
[16]
Siva S, MacManus MP, Martin RF, Martin OA. Abscopal effects of radiation therapy: A clinical review for the radiobiologist. Cancer Lett 2015; 356(1): 82-90.
[17]
Mohammad RM, Muqbil I, Lowe L, et al. Broad targeting of resistance to apoptosis in cancer. Semin Cancer Biol 2015; 35(Suppl.): S78-S103.
[18]
Cree IA, Charlton P. Molecular chess? Hallmarks of anti-cancer drug resistance. BMC Cancer 2017; 17(1): 10.
[19]
Agostinis P, Berg K, Cengel KA, et al. Photodynamic therapy of cancer: An update. CA Cancer J Clin 2011; 61(4): 250-81.
[20]
TriantaphylidA"s C, Havaux M. Singlet oxygen in plants: production, detoxification and signaling. Trends Plant Sci 2009; 14(4): 219-28.
[21]
Onyango AN. Endogenous Generation of Singlet Oxygen and Ozone in Human and Animal Tissues: Mechanisms, Biological Significance, and Influence of Dietary Components. Oxid Med Cell Longev 2016; 2016: 2398573.
[22]
van Straten D, Mashayekhi V, de Bruijn HS, Oliveira S, Robinson DJ. Oncologic Photodynamic Therapy: Basic Principles, Current Clinical Status and Future Directions. Cancers (Basel) 2017; 9(2): E19.
[23]
Borgia F, Giuffrida R, Caradonna E, Vaccaro M, Guarneri F, CannavA SP. Early and Late Onset Side Effects of Photodynamic Therapy. Biomedicines 2018; 6(1): E12.
[24]
Foote CS. Definition of type I and type II photosensitized oxidation. Photochem Photobiol 1991; 54(5): 659.
[25]
Kessel D. The role of low-density lipoprotein in the biodistribution of photosensitizing agents. J Photochem Photobiol B 1992; 14(3): 261-2.
[26]
Lundqvist M, Stigler J, Elia G, Lynch I, Cedervall T, Dawson KA. Nanoparticle size and surface properties determine the protein corona with possible implications for biological impacts. Proc Natl Acad Sci USA 2008; 105(38): 14265-70.
[27]
Kessel D. Correlation between subcellular localization and photodynamic efficacy. J Porphyr Phthalocyanines 2004; 8(08): 1009-14.
[28]
Yeh SC, Diamond KR, Patterson MS, Nie Z, Hayward JE, Fang Q. Monitoring photosensitizer uptake using two photon fluorescence lifetime imaging microscopy. Theranostics 2012; 2(9): 817-26.
[29]
Kou J, Dou D, Yang L. Porphyrin photosensitizers in photodynamic therapy and its applications. Oncotarget 2017; 8(46): 81591-603.
[30]
Maiolino S, Moret F, Conte C, et al. Hyaluronan-decorated polymer nanoparticles targeting the CD44 receptor for the combined photo/chemo-therapy of cancer. Nanoscale 2015; 7(13): 5643-53.
[31]
Huang H, Mallidi S, Obaid G, Sears B, Tangutoori S, Hasan T. Advancing photodynamic therapy with biochemically tuned liposomal nanotechnologies Applications of Nanoscience in Photomedicine 2015; 487-510.
[32]
Buytaert E, Dewaele M, Agostinis P. Molecular effectors of multiple cell death pathways initiated by photodynamic therapy. Biochim Biophys Acta 2007; 1776(1): 86-107.
[33]
Konan YN, Gurny R, All mann E. State of the art in the delivery of photosensitizers for photodynamic therapy. J Photochem Photobiol B 2002; 66(2): 89-106.
[34]
Jeelani S, Reddy RC, Maheswaran T, Asokan GS, Dany A, Anand B. Theranostics: A treasured tailor for tomorrow. J Pharm Bioallied Sci 2014; 6(Suppl. 1): S6-8.
[35]
Wang H, Lv B, Tang Z, et al. Scintillator-Based Nanohybrids with Sacrificial Electron Prodrug for Enhanced X-ray-Induced Photodynamic Therapy. Nano Lett 2018.
[36]
Shen S, Zhu C, Huo D, Yang M, Xue J, Xia Y. A Hybrid Nanomaterial for the Controlled Generation of Free Radicals and Oxidative Destruction of Hypoxic Cancer Cells. Angew Chem Int Ed Engl 2017; 56(30): 8801-4.
[37]
Foote CS. Mechanisms of photosensitized oxidation. There are several different types of photosensitized oxidation which may be important in biological systems. Science 1968; 162(3857): 963-70.
[38]
Dai T, Fuchs BB, Coleman JJ, et al. Concepts and principles of photodynamic therapy as an alternative antifungal discovery platform. Front Microbiol 2012; 3: 120.
[39]
Bacellar IO, Tsubone TM, Pavani C, Baptista MS. Photodynamic Efficiency: From Molecular Photochemistry to Cell Death. Int J Mol Sci 2015; 16(9): 20523-59.
[40]
Magi B, Ettorre A, Liberatori S, et al. Selectivity of protein carbonylation in the apoptotic response to oxidative stress associated with photodynamic therapy: A cell biochemical and proteomic investigation. Cell Death Differ 2004; 11(8): 842-52.
[41]
Roede JR, Jones DP. Reactive species and mitochondrial dysfunction: mechanistic significance of 4-hydroxynonenal. Environ Mol Mutagen 2010; 51(5): 380-90.
[42]
Singh KK, Russell J, Sigala B, Zhang Y, Williams J, Keshav KF. Mitochondrial DNA determines the cellular response to cancer therapeutic agents. Oncogene 1999; 18(48): 6641-6.
[43]
Bauer G. The Antitumor Effect of Singlet Oxygen. Anticancer Res 2016; 36(11): 5649-63.
[44]
Brunelli L, Yermilov V, Beckman JS. Modulation of catalase peroxidatic and catalatic activity by nitric oxide. Free Radic Biol Med 2001; 30(7): 709-14.
[45]
Thiagarajah JR, Chang J, Goettel JA, Verkman AS, Lencer WI. Aquaporin-3 mediates hydrogen peroxide-dependent responses to environmental stress in colonic epithelia. Proc Natl Acad Sci USA 2017; 114(3): 568-73.
[46]
Sies H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: Oxidative eustress. Redox Biol 2017; 11: 613-9.
[47]
Juhasz A, Markel S, Gaur S, et al. NADPH oxidase 1 supports proliferation of colon cancer cells by modulating reactive oxygen species-dependent signal transduction. J Biol Chem 2017; 292(19): 7866-87.
[48]
RiethmA1/4ller M, Burger N, Bauer G. Singlet oxygen treatment of tumor cells triggers extracellular singlet oxygen generation, catalase inactivation and reactivation of intercellular apoptosis-inducing signaling. Redox Biol 2015; 6: 157-68.
[49]
Jung HS, Han J, Shi H, et al. Overcoming the Limits of Hypoxia in Photodynamic Therapy: A Carbonic Anhydrase IX-Targeted Approach. J Am Chem Soc 2017; 139(22): 7595-602.
[50]
Papandreou I, Krishna C, Kaper F, Cai D, Giaccia AJ, Denko NC. Anoxia is necessary for tumor cell toxicity caused by a low-oxygen environment. Cancer Res 2005; 65(8): 3171-8.
[51]
Li C, Jackson RM. Reactive species mechanisms of cellular hypoxia-reoxygenation injury. Am J Physiol Cell Physiol 2002; 282(2): C227-41.
[52]
Stuker F, Ripoll J, Rudin M. Fluorescence molecular tomography: principles and potential for pharmaceutical research. Pharmaceutics 2011; 3(2): 229-74.
[53]
Kim MM, Penjweini R, Gemmell NR, et al. A Comparison of Singlet Oxygen Explicit Dosimetry (SOED) and Singlet Oxygen Luminescence Dosimetry (SOLD) for Photofrin-Mediated Photodynamic Therapy. Cancers (Basel) 2016; 8(12): E109.
[54]
ArdenkjAr-Larsen JH, Fridlund B, Gram A, Hansson G, Hansson L, Lerche MH, et al. Increase in signal-to-noise ratio of > 10,000 times in liquid-state NMR. 2003; 100(18): 10158-63.
[55]
Vasos PR, Comment A, Sarkar R, et al. Long-lived states to sustain hyperpolarized magnetization. Proc Natl Acad Sci USA 2009; 106(44): 18469-73.
[56]
Sandulache VC, Chen Y, Lee J, Rubinstein A, Ramirez MS, Skinner HD, et al. Evaluation of hyperpolarized [1-(1)(3)C]-pyruvate by magnetic resonance to detect ionizing radiation effects in real time. PLoS One 2014; 9(1): e87031.
[57]
Mroz P, Yaroslavsky A, Kharkwal GB, Hamblin MR. Cell death pathways in photodynamic therapy of cancer. Cancers (Basel) 2011; 3(2): 2516-39.
[58]
Galluzzi L, Kepp O, Kroemer G. Enlightening the impact of immunogenic cell death in photodynamic cancer therapy. EMBO J 2012; 31(5): 1055-7.
[59]
Elmore S. Apoptosis: A review of programmed cell death. Toxicol Pathol 2007; 35(4): 495-516.
[60]
Coussens LM, Werb Z. Inflammation and cancer. Nature 2002; 420(6917): 860-7.
[61]
Xue LY, Chiu SM, Oleinick NL. Photochemical destruction of the Bcl-2 oncoprotein during photodynamic therapy with the phthalocyanine photosensitizer Pc 4. Oncogene 2001; 20(26): 3420-7.
[62]
Srivastava M, Ahmad N, Gupta S, Mukhtar H. Involvement of Bcl-2 and Bax in photodynamic therapy-mediated apoptosis. Antisense Bcl-2 oligonucleotide sensitizes RIF 1 cells to photodynamic therapy apoptosis. J Biol Chem 2001; 276(18): 15481-8.
[63]
Guo Q, Dong B, Nan F, Guan D, Zhang Y. 5-Aminolevulinic acid photodynamic therapy in human cervical cancer via the activation of microRNA-143 and suppression of the Bcl-2/Bax signaling pathway. Mol Med Rep 2016; 14(1): 544-50.
[64]
Almeida RD, Manadas BJ, Carvalho AP, Duarte CB. Intracellular signaling mechanisms in photodynamic therapy. Biochim Biophys Acta 2004; 1704(2): 59-86.
[65]
Newton K. RIPK1 and RIPK3: critical regulators of inflammation and cell death. Trends Cell Biol 2015; 25(6): 347-53.
[66]
Zheng L, Bidere N, Staudt D, et al. Competitive control of independent programs of tumor necrosis factor receptor-induced cell death by TRADD and RIP1. Mol Cell Biol 2006; 26(9): 3505-13.
[67]
Fukuyama T, Ichiki Y, Yamada S, et al. Cytokine production of lung cancer cell lines: Correlation between their production and the inflammatory/immunological responses both in vivo and in vitro. Cancer Sci 2007; 98(7): 1048-54.
[68]
Ouyang L, Shi Z, Zhao S, et al. Programmed cell death pathways in cancer: A review of apoptosis, autophagy and programmed necrosis. Cell Prolif 2012; 45(6): 487-98.
[69]
Miki Y, Akimoto J, Moritake K, Hironaka C, Fujiwara Y. Photodynamic therapy using talaporfin sodium induces concentration-dependent programmed necroptosis in human glioblastoma T98G cells. Lasers Med Sci 2015; 30(6): 1739-45.
[70]
Balkwill F. Tumour necrosis factor and cancer. Nat Rev Cancer 2009; 9(5): 361-71.
[71]
Nakajima S, Kitamura M. Bidirectional regulation of NF-IB by reactive oxygen species: A role of unfolded protein response. Free Radic Biol Med 2013; 65: 162-74.
[72]
Broekgaarden M, Weijer R, van Gulik TM, Hamblin MR, Heger M. Tumor cell survival pathways activated by photodynamic therapy: A molecular basis for pharmacological inhibition strategies. Cancer Metastasis Rev 2015; 34(4): 643-90.
[73]
Piette J. Signalling pathway activation by photodynamic therapy: NF-IB at the crossroad between oncology and immunology. Photochem Photobiol Sci 2015; 14(8): 1510-7.
[74]
Wang CY, Mayo MW, Baldwin AS Jr. TNF- and cancer therapy-induced apoptosis: potentiation by inhibition of NF-kappaB. Science 1996; 274(5288): 784-7.
[75]
Coupienne I, Bontems S, Dewaele M, et al. NF-kappaB inhibition improves the sensitivity of human glioblastoma cells to 5-aminolevulinic acid-based photodynamic therapy. Biochem Pharmacol 2011; 81(5): 606-16.
[76]
Kearney CJ, Martin SJ. An Inflammatory Perspective on Necroptosis. Mol Cell 2017; 65(6): 965-73.
[77]
Tsubone TM, Martins WK, Pavani C, Junqueira HC, Itri R, Baptista MS. Enhanced efficiency of cell death by lysosome-specific photodamage. Sci Rep 2017; 7(1): 6734.
[78]
Aits S, JA M. Lysosomal cell death at a glance. J Cell Sci 2013; 126(Pt 9): 1905-12.
[79]
Repnik U, Stoka V, Turk V, Turk B. Lysosomes and lysosomal cathepsins in cell death. Biochim Biophys Acta 2012; 1824(1): 22-33.
[80]
Kessel D, Luo Y, Mathieu P, Reiners JJ Jr. Determinants of the apoptotic response to lysosomal photodamage. Photochem Photobiol 2000; 71(2): 196-200.
[81]
Wang F, Salvati A, Boya P. Lysosome-dependent cell death and deregulated autophagy induced by amine-modified polystyrene nanoparticles. Open Biol 2018; 8(4): 170271.
[82]
Kav? N, Pegan K, Turk B. Lysosomes in programmed cell death pathways: from initiators to amplifiers. Biol Chem 2016; 398(3): 289-301.
[83]
Berg K, Moan J. Lysosomes as photochemical targets. Int J Cancer 1994; 59(6): 814-22.
[84]
Reiners JJ Jr, Caruso JA, Mathieu P, Chelladurai B, Yin XM, Kessel D. Release of cytochrome c and activation of pro-caspase-9 following lysosomal photodamage involves Bid cleavage. Cell Death Differ 2002; 9(9): 934-44.
[85]
Papadopoulos C, Meyer H. Detection and Clearance of Damaged Lysosomes by the Endo-Lysosomal Damage Response and Lysophagy. Curr Biol 2017; 27(24): R1330-41.
[86]
BAegyi G, Baumeister P, Benedetti A, et al. Endoplasmic reticulum stress. Ann N Y Acad Sci 2007; 1113: 58-71.
[87]
Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol 2007; 8(7): 519-29.
[88]
Moserova I, Kralova J. Role of ER stress response in photodynamic therapy: ROS generated in different subcellular compartments trigger diverse cell death pathways. PLoS One 2012; 7(3): e32972.
[89]
Szegezdi E, Logue SE, Gorman AM, Samali A. Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO Rep 2006; 7(9): 880-5.
[90]
Dandekar A, Mendez R, Zhang K. Cross talk between ER stress, oxidative stress, and inflammation in health and disease. Methods Mol Biol 2015; 1292: 205-14.
[91]
Wang M, Kaufman RJ. The impact of the endoplasmic reticulum protein-folding environment on cancer development. Nat Rev Cancer 2014; 14(9): 581-97.
[92]
Verfaillie T, Rubio N, Garg AD, et al. PERK is required at the ER-mitochondrial contact sites to convey apoptosis after ROS-based ER stress. Cell Death Differ 2012; 19(11): 1880-91.
[93]
Li G, Scull C, Ozcan L, Tabas I. NADPH oxidase links endoplasmic reticulum stress, oxidative stress, and PKR activation to induce apoptosis. J Cell Biol 2010; 191(6): 1113-25.
[94]
Rao RV, Ellerby HM, Bredesen DE. Coupling endoplasmic reticulum stress to the cell death program. Cell Death Differ 2004; 11(4): 372-80.
[95]
PArn-Ares MI, Samali A, Orrenius S. Cleavage of the calpain inhibitor, calpastatin, during apoptosis. Cell Death Differ 1998; 5(12): 1028-33.
[96]
SchrAder M. Endoplasmic reticulum stress responses. Cell Mol Life Sci 2008; 65(6): 862-94.
[97]
Grimm S. The ER-mitochondria interface: the social network of cell death. Biochim Biophys Acta 2012; 1823(2): 327-34.
[98]
Ghibelli L, Grzanka A. Organelle cross-talk in apoptotic and survival pathways. Int J Cell Biol 2012; 2012: 968586.
[99]
Dikic I, Elazar Z. Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol 2018; 19(6): 349-64.
[100]
Kessel D, Arroyo AS. Apoptotic and autophagic responses to Bcl-2 inhibition and photodamage. Photochem Photobiol Sci 2007; 6(12): 1290-5.
[101]
Reiners JJ Jr, Agostinis P, Berg K, Oleinick NL, Kessel D. Assessing autophagy in the context of photodynamic therapy. Autophagy 2010; 6(1): 7-18.
[102]
Buytaert E, Callewaert G, Hendrickx N, et al. Role of endoplasmic reticulum depletion and multidomain proapoptotic BAX and BAK proteins in shaping cell death after hypericin-mediated photodynamic therapy. FASEB J 2006; 20(6): 756-8.
[103]
Kessel D, Reiners JJ Jr. Promotion of Proapoptotic Signals by Lysosomal Photodamage. Photochem Photobiol 2015; 91(4): 931-6.
[104]
Acedo P, Stockert JC, CaAete M, Villanueva A. Two combined photosensitizers: A goal for more effective photodynamic therapy of cancer. Cell Death Dis 2014; 5: e1122.
[105]
Villanueva A, Stockert JC, CaAte M, Acedo P. A new protocol in photodynamic therapy: enhanced tumour cell death by combining two different photosensitizers. Photochem Photobiol Sci 2010; 9(3): 295-7.
[106]
Liu J, Wang Z. Increased Oxidative Stress as a Selective Anticancer Therapy. Oxid Med Cell Longev 2015; 2015: 294303.
[107]
Wang GD, Nguyen HT, Chen H, et al. X-Ray Induced Photodynamic Therapy: A Combination of Radiotherapy and Photodynamic Therapy. Theranostics 2016; 6(13): 2295-305.
[108]
Moan J, Berg K. The photodegradation of porphyrins in cells can be used to estimate the lifetime of singlet oxygen. Photochem Photobiol 1991; 53(4): 549-53.
[109]
Finkel T. Signal transduction by reactive oxygen species. J Cell Biol 2011; 194(1): 7-15.
[110]
Dahle J, Bagdonas S, Kaalhus O, Olsen G, Steen HB, Moan J. The bystander effect in photodynamic inactivation of cells. Biochim Biophys Acta 2000; 1475(3): 273-80.
[111]
Azzam EI, Little JB. The radiation-induced bystander effect: evidence and significance. Hum Exp Toxicol 2004; 23(2): 61-5.
[112]
Mothersill C, Seymour CB. Radiation-induced bystander effects--implications for cancer. Nat Rev Cancer 2004; 4(2): 158-64.
[113]
de la Torre Gomez C, Goreham RV, Bech Serra JJ, Nann T, Kussmann M. “Exosomics”-A Review of Biophysics, Biology and Biochemistry of Exosomes With a Focus on Human Breast Milk. Front Genet 2018; 9: 92.
[114]
Sun W, Luo JD, Jiang H, Duan DD. Tumor exosomes: A double-edged sword in cancer therapy. Acta Pharmacol Sin 2018; 39(4): 534-41.
[115]
Aubertin K, Silva AK, Luciani N, et al. Massive release of extracellular vesicles from cancer cells after photodynamic treatment or chemotherapy. Sci Rep 2016; 6: 35376.
[116]
Theodoraki MN, Yerneni SS, Brunner C, Theodorakis J, Hoffmann TK, Whiteside TL. Plasma-derived Exosomes Reverse Epithelial-to-Mesenchymal Transition after Photodynamic Therapy of Patients with Head and Neck Cancer. Oncoscience 2018; 5(3-4): 75-87.
[117]
Tschopp J, Schroder K. NLRP3 inflammasome activation: The convergence of multiple signalling pathways on ROS production? Nat Rev Immunol 2010; 10(3): 210-5.
[118]
Matzinger P. The danger model: A renewed sense of self. Science 2002; 296(5566): 301-5.
[119]
Garg AD, Agostinis P. Cell death and immunity in cancer: From danger signals to mimicry of pathogen defense responses. Immunol Rev 2017; 280(1): 126-48.
[120]
Zhang Q, Zhu B, Li Y. Resolution of Cancer-Promoting Inflammation: A New Approach for Anticancer Therapy. Front Immunol 2017; 8: 71.
[121]
Duo CC, Gong FY, He XY, et al. Soluble calreticulin induces tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 production by macrophages through mitogen-activated protein kinase (MAPK) and NFI signaling pathways. Int J Mol Sci 2014; 15(2): 2916-28.
[122]
Golden EB, Formenti SC. Radiation therapy and immunotherapy: growing pains. Int J Radiat Oncol Biol Phys 2015; 91(2): 252-4.
[123]
Golden EB, Chhabra A, Chachoua A, et al. Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: A proof-of-principle trial. Lancet Oncol 2015; 16(7): 795-803.
[124]
Ng J, Dai T. Radiation therapy and the abscopal effect: A concept comes of age. Ann Transl Med 2016; 4(6): 118.
[125]
Tang C, Wang X, Soh H, et al. Combining radiation and immunotherapy: A new systemic therapy for solid tumors? Cancer Immunol Res 2014; 2(9): 831-8.
[126]
Garg AD, Dudek-Peric AM, Romano E, Agostinis P. Immunogenic cell death. Int J Dev Biol 2015; 59(1-3): 131-40.
[127]
Panzarini E, Inguscio V, Dini L. Immunogenic cell death: can it be exploited in PhotoDynamic Therapy for cancer? BioMed Res Int 2013; 2013: 482160.
[128]
Korbelik M, Sun J, Cecic I. Photodynamic therapy-induced cell surface expression and release of heat shock proteins: relevance for tumor response. Cancer Res 2005; 65(3): 1018-26.
[129]
Etminan N, Peters C, Lakbir D, et al. Heat-shock protein 70-dependent dendritic cell activation by 5-aminolevulinic acid-mediated photodynamic treatment of human glioblastoma spheroids in vitro. Br J Cancer 2011; 105(7): 961-9.
[130]
Korbelik M, Sun J. Photodynamic therapy-generated vaccine for cancer therapy. Cancer Immunol Immunother 2006; 55(8): 900-9.
[131]
Chen GY, NuAez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol 2010; 10(12): 826-37.
[132]
Korbelik M. Cancer vaccines generated by photodynamic therapy. Photochem Photobiol Sci 2011; 10(5): 664-9.
[133]
Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol 2010; 28: 367-88.
[134]
Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 2002; 418(6894): 191-5.
[135]
He S, Cheng J, Sun L, et al. HMGB1 released by irradiated tumor cells promotes living tumor cell proliferation via paracrine effect. Cell Death Dis 2018; 9(6): 648.
[136]
Kazama H, Ricci JE, Herndon JM, Hoppe G, Green DR, Ferguson TA. Induction of immunological tolerance by apoptotic cells requires caspase-dependent oxidation of high-mobility group box-1 protein. Immunity 2008; 29(1): 21-32.
[137]
Tang D, Kang R, Cheh CW, et al. HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene 2010; 29(38): 5299-310.
[138]
Garg AD, Krysko DV, Verfaillie T, et al. A novel pathway combining calreticulin exposure and ATP secretion in immunogenic cancer cell death. EMBO J 2012; 31(5): 1062-79.
[139]
Garg AD, Dudek AM, Ferreira GB, et al. ROS-induced autophagy in cancer cells assists in evasion from determinants of immunogenic cell death. Autophagy 2013; 9(9): 1292-307.
[140]
Dudek AM, Garg AD, Krysko DV, De Ruysscher D, Agostinis P. Inducers of immunogenic cancer cell death. Cytokine Growth Factor Rev 2013; 24(4): 319-33.
[141]
Du HY, Olivo M, Mahendran R, et al. Hypericin photoactivation triggers down-regulation of matrix metalloproteinase-9 expression in well-differentiated human nasopharyngeal cancer cells. Cell Mol Life Sci 2007; 64(7-8): 979-88.
[142]
Gollnick SO, Brackett CM. Enhancement of anti-tumor immunity by photodynamic therapy. Immunol Res 2010; 46(1-3): 216-26.
[143]
Matroule JY, Bonizzi G, MorliA"re P, et al. Pyropheophorbide-a methyl ester-mediated photosensitization activates transcription factor NF-kappaB through the interleukin-1 receptor-dependent signaling pathway. J Biol Chem 1999; 274(5): 2988-3000.
[144]
Kick G, Messer G, Goetz A, Plewig G, Kind P. Photodynamic therapy induces expression of interleukin 6 by activation of AP-1 but not NF-kappa B DNA binding. Cancer Res 1995; 55(11): 2373-9.
[145]
Casbon AJ, Reynaud D, Park C, et al. Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils. Proc Natl Acad Sci USA 2015; 112(6): E566-75.
[146]
Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol 2007; 25: 267-96.
[147]
Spary LK, Salimu J, Webber JP, Clayton A, Mason MD, Tabi Z. Tumor stroma-derived factors skew monocyte to dendritic cell differentiation toward a suppressive CD14+ PD-L1+ phenotype in prostate cancer. OncoImmunology 2014; 3(9): e955331.
[148]
Ghirelli C, Hagemann T. Targeting immunosuppression for cancer therapy. J Clin Invest 2013; 123(6): 2355-7.
[149]
Carta S, Castellani P, Delfino L, Tassi S, VenA" R, Rubartelli A. DAMPs and inflammatory processes: the role of redox in the different outcomes. J Leukoc Biol 2009; 86(3): 549-55.
[150]
Hernandez C, Huebener P, Schwabe RF. Damage-associated molecular patterns in cancer: A double-edged sword. Oncogene 2016; 35(46): 5931-41.
[151]
Mitra S, Foster TH. In vivo confocal fluorescence imaging of the intratumor distribution of the photosensitizer mono-L-aspartylchlorin-e6. Neoplasia 2008; 10(5): 429-38.
[152]
Fucikova J, Moserova I, Urbanova L, et al. Prognostic and Predictive Value of DAMPs and DAMP-Associated Processes in Cancer. Front Immunol 2015; 6: 402.
[153]
Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015; 348(6230): 56-61.
[154]
Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 2015; 15(8): 486-99.
[155]
Galluzzi L, Vacchelli E, Bravo-San Pedro JM, et al. Classification of current anticancer immunotherapies. Oncotarget 2014; 5(24): 12472-508.
[156]
Korbelik M, Zhang W, Merchant S. Involvement of damage-associated molecular patterns in tumor response to photodynamic therapy: surface expression of calreticulin and high-mobility group box-1 release. Cancer Immunol Immunother 2011; 60(10): 1431-7.
[157]
Korbelik M. Impact of cell death manipulation on the efficacy of photodynamic therapy-generated cancer vaccines. World J Immunol 2015; 5(3): 95-8.
[158]
Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med 2010; 49(11): 1603-16.
[159]
Plaks V, Kong N, Werb Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 2015; 16(3): 225-38.
[160]
Swanton C. Intratumor heterogeneity: evolution through space and time. Cancer Res 2012; 72(19): 4875-82.
[161]
Gorrini C, Harris IS, Mak TW. Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov 2013; 12(12): 931-47.
[162]
Ma Q. Role of nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol 2013; 53: 401-26.
[163]
Nguyen T, Sherratt PJ, Nioi P, Yang CS, Pickett CB. Nrf2 controls constitutive and inducible expression of ARE-driven genes through a dynamic pathway involving nucleocytoplasmic shuttling by Keap1. J Biol Chem 2005; 280(37): 32485-92.
[164]
Pajares M, Jim nez-Moreno N, Dias IH, et al. Redox control of protein degradation. Redox Biol 2015; 6: 409-20.
[165]
Hayes JD, Dinkova-Kostova AT. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem Sci 2014; 39(4): 199-218.
[166]
Panieri E, Santoro MM. ROS homeostasis and metabolism: A dangerous liason in cancer cells. Cell Death Dis 2016; 7(6): e2253.
[167]
Schumacker PT. Reactive oxygen species in cancer cells: live by the sword, die by the sword. Cancer Cell 2006; 10(3): 175-6.
[168]
Ryoo IG, Lee SH, Kwak MK. Redox Modulating NRF2: A Potential Mediator of Cancer Stem Cell Resistance. Oxid Med Cell Longev 2016; 2016: 2428153.
[169]
Emmink BL, Verheem A, Van Houdt WJ, et al. The secretome of colon cancer stem cells contains drug-metabolizing enzymes. J Proteomics 2013; 91: 84-96.
[170]
DeNicola GM, Karreth FA, Humpton TJ, et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature 2011; 475(7354): 106-9.
[171]
Mitsuishi Y, Taguchi K, Kawatani Y, et al. Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell 2012; 22(1): 66-79.
[172]
Rojo AI, Rada P, Mendiola M, et al. The PTEN/NRF2 axis promotes human carcinogenesis. Antioxid Redox Signal 2014; 21(18): 2498-514.
[173]
Eades G, Yang M, Yao Y, Zhang Y, Zhou Q. miR-200a regulates Nrf2 activation by targeting Keap1 mRNA in breast cancer cells. J Biol Chem 2011; 286(47): 40725-33.
[174]
Kim YR, Oh JE, Kim MS, et al. Oncogenic NRF2 mutations in squamous cell carcinomas of oesophagus and skin. J Pathol 2010; 220(4): 446-51.
[175]
Shibata T, Kokubu A, Saito S, et al. NRF2 mutation confers malignant potential and resistance to chemoradiation therapy in advanced esophageal squamous cancer. Neoplasia 2011; 13(9): 864-73.
[176]
Singh A, Misra V, Thimmulappa RK, et al. Dysfunctional KEAP1-NRF2 interaction in non-small-cell lung cancer. PLoS Med 2006; 3(10): e420.
[177]
Kerins MJ, Ooi A. A catalogue of somatic NRF2 gain-of-function mutations in cancer. Sci Rep 2018; 8(1): 12846.
[178]
McMahon M, Itoh K, Yamamoto M, Hayes JD. Keap1-dependent proteasomal degradation of transcription factor Nrf2 contributes to the negative regulation of antioxidant response element-driven gene expression. J Biol Chem 2003; 278(24): 21592-600.
[179]
Cuadrado A. 2015.
[180]
Rada P, Rojo AI, Chowdhry S, McMahon M, Hayes JD, Cuadrado A. SCF/beta-TrCP promotes glycogen synthase kinase 3-dependent degradation of the Nrf2 transcription factor in a Keap1-independent manner. Mol Cell Biol 2011; 31(6): 1121-33.
[181]
Rada P, Rojo AI, Evrard-Todeschi N, et al. Structural and functional characterization of Nrf2 degradation by the glycogen synthase kinase 3/I-TrCP axis. Mol Cell Biol 2012; 32(17): 3486-99.
[182]
Taguchi K, Motohashi H, Yamamoto M. Molecular mechanisms of the Keap1?"Nrf2 pathway in stress response and cancer evolution. Genes Cells 2011; 16(2): 123-40.
[183]
Chen W, Sun Z, Wang XJ, et al. Direct interaction between Nrf2 and p21(Cip1/WAF1) upregulates the Nrf2-mediated antioxidant response. Mol Cell 2009; 34(6): 663-73.
[184]
Katsuragi Y, Ichimura Y, Komatsu M. Regulation of the Keap1"Nrf2 pathway by p62/SQSTM1. Curr Opin Toxicol 2016; 1: 54-61.
[185]
Liu WJ, Ye L, Huang WF, et al. p62 links the autophagy pathway and the ubiqutin-proteasome system upon ubiquitinated protein degradation. Cell Mol Biol Lett 2016; 21: 29.
[186]
Pajares M, Jim nez-Moreno N, GarcA-a-Yage AJ, et al. Transcription factor NFE2L2/NRF2 is a regulator of macroautophagy genes. Autophagy 2016; 12(10): 1902-16.
[187]
Pajares M, Rojo AI, Arias E, DA-az-Carretero A, Cuervo AM, Cuadrado A. Transcription factor NFE2L2/NRF2 modulates chaperone-mediated autophagy through the regulation of LAMP2A. Autophagy 2018; 14(8): 1310-22.
[188]
Motohashi H, Katsuoka F, Engel JD, Yamamoto M. Small Maf proteins serve as transcriptional cofactors for keratinocyte differentiation in the Keap1-Nrf2 regulatory pathway. Proc Natl Acad Sci USA 2004; 101(17): 6379-84.
[189]
Dhakshinamoorthy S, Jain AK, Bloom DA, Jaiswal AK. Bach1 competes with Nrf2 leading to negative regulation of the antioxidant response element (ARE)-mediated NAD(P)H:quinone oxidoreductase 1 gene expression and induction in response to antioxidants. J Biol Chem 2005; 280(17): 16891-900.
[190]
Ziady AG, Sokolow A, Shank S, et al. Interaction with CREB binding protein modulates the activities of Nrf2 and NF-IB in cystic fibrosis airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2012; 302(11): L1221-31.
[191]
Kobayashi EH, Suzuki T, Funayama R, et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nat Commun 2016; 7: 11624.
[192]
Beury DW, Carter KA, Nelson C, et al. Myeloid-Derived Suppressor Cell Survival and Function Are Regulated by the Transcription Factor Nrf2. J Immunol 2016; 196(8): 3470-8.
[193]
Kansanen E, Kuosmanen SM, Leinonen H, Levonen AL. The Keap1-Nrf2 pathway: Mechanisms of activation and dysregulation in cancer. Redox Biol 2013; 1: 45-9.
[194]
Milkovic L, Zarkovic N, Saso L. Controversy about pharmacological modulation of Nrf2 for cancer therapy. Redox Biol 2017; 12: 727-32.
[195]
Rojo de la Vega M, Chapman E, Zhang DD. NRF2 and the Hallmarks of Cancer. Cancer Cell 2018; 34(1): 21-43.
[196]
Trachootham D, Alexandre J, Huang P. Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach? Nat Rev Drug Discov 2009; 8(7): 579-91.
[197]
Klotz LO, SAnchez-Ramos C, Prieto-Arroyo I, UrbAnek P, Steinbrenner H, Monsalve M. Redox regulation of FoxO transcription factors. Redox Biol 2015; 6: 51-72.
[198]
Truong TH, Carroll KS. Redox regulation of epidermal growth factor receptor signaling through cysteine oxidation. Biochemistry 2012; 51(50): 9954-65.
[199]
Becks L, Prince M, Burson H, et al. Aggressive mammary carcinoma progression in Nrf2 knockout mice treated with 7,12-dimethylbenz[a]anthracene. BMC Cancer 2010; 10: 540.
[200]
Schmidt HH, Stocker R, Vollbracht C, et al. Antioxidants in Translational Medicine. Antioxid Redox Signal 2015; 23(14): 1130-43.
[201]
BarabAsi AL, Gulbahce N, Loscalzo J. Network medicine: A network-based approach to human disease. Nat Rev Genet 2011; 12(1): 56-68.
[202]
Poornima P, Kumar JD, Zhao Q, Blunder M, Efferth T. Network pharmacology of cancer: From understanding of complex interactomes to the design of multi-target specific therapeutics from nature. Pharmacol Res 2016; 111: 290-302.
[203]
Bomprezzi R. Dimethyl fumarate in the treatment of relapsing-remitting multiple sclerosis: An overview. Ther Adv Neurol Disorder 2015; 8(1): 20-30.
[204]
Dao VT, Casas AI, Maghzal GJ, et al. Pharmacology and Clinical Drug Candidates in Redox Medicine. Antioxid Redox Signal 2015; 23(14): 1113-29.
[205]
Kitamura H, Motohashi H. NRF2 addiction in cancer cells. Cancer Sci 2018; 109(4): 900-11.
[206]
Okano Y, Nezu U, Enokida Y, et al. SNP (-617C>A) in ARE-like loci of the NRF2 gene: A new biomarker for prognosis of lung adenocarcinoma in Japanese non-smoking women. PLoS One 2013; 8(9): e73794.
[207]
Ishikawa T, Kajimoto Y, Sun W, et al. Role of Nrf2 in cancer photodynamic therapy: regulation of human ABC transporter ABCG2. J Pharm Sci 2013; 102(9): 3058-69.
[208]
Zhou S, Ye W, Shao Q, Zhang M, Liang J. Nrf2 is a potential therapeutic target in radioresistance in human cancer. Crit Rev Oncol Hematol 2013; 88(3): 706-15.
[209]
Rojo de la Vega M, Dodson M, Chapman E, Zhang DD. NRF2-targeted therapeutics: New targets and modes of NRF2 regulation. Curr Opin Toxicol 2016; 1: 62-70.
[210]
Singh A, Venkannagari S, Oh KH, et al. Small Molecule Inhibitor of NRF2 Selectively Intervenes Therapeutic Resistance in KEAP1-Deficient NSCLC Tumors. ACS Chem Biol 2016; 11(11): 3214-25.
[211]
Olayanju A, Copple IM, Bryan HK, et al. Brusatol provokes a rapid and transient inhibition of Nrf2 signaling and sensitizes mammalian cells to chemical toxicity-implications for therapeutic targeting of Nrf2. Free Radic Biol Med 2015; 78: 202-12.
[212]
Sun X, Wang Q, Wang Y, Du L, Xu C, Liu Q. Brusatol Enhances the Radiosensitivity of A549 Cells by Promoting ROS Production and Enhancing DNA Damage. Int J Mol Sci 2016; 17(7): E997.
[213]
Vartanian S, Ma TP, Lee J, et al. Application of Mass Spectrometry Profiling to Establish Brusatol as an Inhibitor of Global Protein Synthesis. Mol Cell Proteomics 2016; 15(4): 1220-31.
[214]
Wang XJ, Hayes JD, Henderson CJ, Wolf CR. Identification of retinoic acid as an inhibitor of transcription factor Nrf2 through activation of retinoic acid receptor alpha. Proc Natl Acad Sci USA 2007; 104(49): 19589-94.
[215]
Magesh S, Chen Y, Hu L. Small molecule modulators of Keap1-Nrf2-ARE pathway as potential preventive and therapeutic agents. Med Res Rev 2012; 32(4): 687-726.
[216]
Manna A, De Sarkar S, De S, Bauri AK, Chattopadhyay S, Chatterjee M. The variable chemotherapeutic response of Malabaricone-A in leukemic and solid tumor cell lines depends on the degree of redox imbalance. Phytomedicine 2015; 22(7-8): 713-23.
[217]
Zhong H, Xiao M, Zarkovic K, et al. Mitochondrial control of apoptosis through modulation of cardiolipin oxidation in hepatocellular carcinoma: A novel link between oxidative stress and cancer. Free Radic Biol Med 2017; 102: 67-76.
[218]
Wang X, Campos CR, Peart JC, et al. Nrf2 upregulates ATP binding cassette transporter expression and activity at the blood-brain and blood-spinal cord barriers. J Neurosci 2014; 34(25): 8585-93.
[219]
Gao AM, Ke ZP, Wang JN, Yang JY, Chen SY, Chen H. Apigenin sensitizes doxorubicin-resistant hepatocellular carcinoma BEL-7402/ADM cells to doxorubicin via inhibiting PI3K/Akt/Nrf2 pathway. Carcinogenesis 2013; 34(8): 1806-14.
[220]
Lamberti MJ, Vittar NB, da Silva Fde C, Ferreira VF, Rivarola VA. Synergistic enhancement of antitumor effect of I-Lapachone by photodynamic induction of quinone oxidoreductase (NQO1). Phytomedicine 2013; 20(11): 1007-12.
[221]
Hagiya Y, Adachi T, Ogura S, et al. Nrf2-dependent induction of human ABC transporter ABCG2 and heme oxygenase-1 in HepG2 cells by photoactivation of porphyrins: biochemical implications for cancer cell response to photodynamic therapy. J Exp Ther Oncol 2008; 7(2): 153-67.
[222]
Choi BH, Ryoo IG, Kang HC, Kwak MK. The sensitivity of cancer cells to pheophorbide a-based photodynamic therapy is enhanced by Nrf2 silencing. PLoS One 2014; 9(9): e107158.
[223]
Kocanova S, Buytaert E, Matroule JY, et al. Induction of heme-oxygenase 1 requires the p38MAPK and PI3K pathways and suppresses apoptotic cell death following hypericin-mediated photodynamic therapy. Apoptosis 2007; 12(4): 731-41.
[224]
Masoud GN, Li W. HIF-1I pathway: role, regulation and intervention for cancer therapy. Acta Pharm Sin B 2015; 5(5): 378-89.
[225]
Reichard JF, Sartor MA, Puga A. BACH1 is a specific repressor of HMOX1 that is inactivated by arsenite. J Biol Chem 2008; 283(33): 22363-70.
[226]
Kapitulnik J. Bilirubin: An endogenous product of heme degradation with both cytotoxic and cytoprotective properties. Mol Pharmacol 2004; 66(4): 773-9.
[227]
Kapitulnik J, Maines MD. Pleiotropic functions of biliverdin reductase: cellular signaling and generation of cytoprotective and cytotoxic bilirubin. Trends Pharmacol Sci 2009; 30(3): 129-37.
[228]
Dudnik LB, Khrapova NG. Characterization of bilirubin inhibitory properties in free radical oxidation reactions. Membr Cell Biol 1998; 12(2): 233-40.
[229]
Bauer M, Bauer I. Heme oxygenase-1: redox regulation and role in the hepatic response to oxidative stress. Antioxid Redox Signal 2002; 4(5): 749-58.
[230]
Ishikawa T, Nakagawa H, Hagiya Y, Nonoguchi N, Miyatake S, Kuroiwa T. Key Role of Human ABC Transporter ABCG2 in Photodynamic Therapy and Photodynamic Diagnosis. Adv Pharmacol Sci 2010; 2010: 587306.
[231]
Singh A, Wu H, Zhang P, Happel C, Ma J, Biswal S. Expression of ABCG2 (BCRP) is regulated by Nrf2 in cancer cells that confers side population and chemoresistance phenotype. Mol Cancer Ther 2010; 9(8): 2365-76.
[232]
Liu W, Baer MR, Bowman MJ, et al. The tyrosine kinase inhibitor imatinib mesylate enhances the efficacy of photodynamic therapy by inhibiting ABCG2. Clin Cancer Res 2007; 13(8): 2463-70.
[233]
Lee SJ, Hwang HJ, Shin JI, Ahn JC, Chung PS. Enhancement of cytotoxic effect on human head and neck cancer cells by combination of photodynamic therapy and sulforaphane. Gen Physiol Biophys 2015; 34(1): 13-21.
[234]
Mikolajewska P, Juzeniene A, Moan J. Effect of (R)L-sulforaphane on 5-aminolevulinic acid-mediated photodynamic therapy. Transl Res 2008; 152(3): 128-33.
[235]
Kaczy"ska A, Herman-Antosiewicz A. Combination of lapatinib with isothiocyanates overcomes drug resistance and inhibits migration of HER2 positive breast cancer cells. Breast Cancer 2017; 24(2): 271-80.
[236]
Pawlik A, Wiczk A, Kaczy"ska A, Antosiewicz J, Herman-Antosiewicz A. Sulforaphane inhibits growth of phenotypically different breast cancer cells. Eur J Nutr 2013; 52(8): 1949-58.
[237]
Sakao K, Singh SVD. D,L-sulforaphane-induced apoptosis in human breast cancer cells is regulated by the adapter protein p66Shc. J Cell Biochem 2012; 113(2): 599-610.
[238]
Bennett Saidu NE, Bretagne M, Mansuet AL, et al. Dimethyl fumarate is highly cytotoxic in KRAS mutated cancer cells but spares non-tumorigenic cells. Oncotarget 2018; 9(10): 9088-99.
[239]
Theodossiou TA, Olsen CE, Jonsson M, Kubin A, Hothersall JS, Berg K. The diverse roles of glutathione-associated cell resistance against hypericin photodynamic therapy. Redox Biol 2017; 12: 191-7.
[240]
Yang H, Magilnick N, Lee C, et al. Nrf1 and Nrf2 regulate rat glutamate-cysteine ligase catalytic subunit transcription indirectly via NF-kappaB and AP-1. Mol Cell Biol 2005; 25(14): 5933-46.
[241]
Abrahamse H, Kruger CA, Kadanyo S, Mishra A. Nanoparticles for Advanced Photodynamic Therapy of Cancer. Photomed Laser Surg 2017; 35(11): 581-8.
[242]
Calixto GM, Bernegossi J, de Freitas LM, Fontana CR, Chorilli M. Nanotechnology-Based Drug Delivery Systems for Photodynamic Therapy of Cancer: A Review. Molecules 2016; 21(3): 342.
[243]
Kotagiri N, Sudlow GP, Akers WJ, Achilefu S. Breaking the depth dependency of phototherapy with Cerenkov radiation and low-radiance-responsive nanophotosensitizers. Nat Nanotechnol 2015; 10(4): 370-9.
[244]
Yuzhakova DV, Lermontova SA, Grigoryev IS, et al. In vivo multimodal tumor imaging and photodynamic therapy with novel theranostic agents based on the porphyrazine framework-chelated gadolinium (III) cation. Biochim Biophys Acta, Gen Subj 2017; 1861(12): 3120-30.
[245]
Jarvi MT, Niedre MJ, Patterson MS, Wilson BC. Singlet oxygen luminescence dosimetry (SOLD) for photodynamic therapy: current status, challenges and future prospects. Photochem Photobiol 2006; 82(5): 1198-210.
[246]
Kareliotis G, Liossi S, Makropoulou M. Assessment of singlet oxygen dosimetry concepts in photodynamic therapy through computational modeling. Photodiagn Photodyn Ther 2018; 21: 224-33.
[247]
Quirk BJ, Brandal G, Donlon S, et al. Photodynamic therapy (PDT) for malignant brain tumors--where do we stand? Photodiagn Photodyn Ther 2015; 12(3): 530-44.
[248]
Kamkaew A, Chen F, Zhan Y, Majewski RL, Cai W. Scintillating Nanoparticles as Energy Mediators for Enhanced Photodynamic Therapy. ACS Nano 2016; 10(4): 3918-35.
[249]
Babincová M, Sourivong P, Babinec P. Gene transfer-mediated intracellular photodynamic therapy. Med Hypotheses 2000; 52(2): 180-.
[250]
Sadanala KC, Chaturvedi PK, Seo YM, et al. Sono-photodynamic combination therapy: A review on sensitizers. Anticancer Res 2014; 34(9): 4657-64.
[251]
Guo Y, Sheng S, Zhang W, Lun M, Tsai S-M, Chin W-C, Eds. High energy photons excited photodynamic cancer therapy in vitro Optical Methods for Tumor Treatment and Detection: Mechanisms and Techniques in Photodynamic Therapy XXVII 2018.
[252]
Ai F, Ju Q, Zhang X, Chen X, Wang F, Zhu G. A core-shell-shell nanoplatform upconverting near-infrared light at 808 nm for luminescence imaging and photodynamic therapy of cancer. Sci Rep 2015; 5: 10785.
[253]
Akimoto J. Photodynamic Therapy for Malignant Brain Tumors. Neurol Med Chir (Tokyo) 2016; 56(4): 151-7.
[254]
Salem A, Asselin MC, Reymen B, et al. Targeting Hypoxia to Improve Non-Small Cell Lung Cancer Outcome. J Natl Cancer Inst 2018; 110(1)
[255]
Mitra S, Foster TH. Carbogen breathing significantly enhances the penetration of red light in murine tumours in vivo. Phys Med Biol 2004; 49(10): 1891-904.
[256]
Chen B, Roskams T, de Witte PA. Antivascular tumor eradication by hypericin-mediated photodynamic therapy. Photochem Photobiol 2002; 76(5): 509-13.
[257]
Elliott MR, Chekeni FB, Trampont PC, et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature 2009; 461(7261): 282-6.
[258]
Amores-Iniesta J, BarberA -Cremades M, MartA-nez CM, et al. Extracellular ATP Activates the NLRP3 Inflammasome and Is an Early Danger Signal of Skin Allograft Rejection. Cell Reports 2017; 21(12): 3414-26.
[259]
Cruz CM, Rinna A, Forman HJ, Ventura AL, Persechini PM, Ojcius DM. ATP activates a reactive oxygen species-dependent oxidative stress response and secretion of proinflammatory cytokines in macrophages. J Biol Chem 2007; 282(5): 2871-9.
[260]
Menu P, Mayor A, Zhou R, et al. ER stress activates the NLRP3 inflammasome via an UPR-independent pathway. Cell Death Dis 2012; 3: e261.
[261]
Abais JM, Xia M, Zhang Y, Boini KM, Li PL. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxid Redox Signal 2015; 22(13): 1111-29.
[262]
Shi Y, Zheng W, Rock KL. Cell injury releases endogenous adjuvants that stimulate cytotoxic T cell responses. Proc Natl Acad Sci USA 2000; 97(26): 14590-5.
[263]
Dostert C, P trilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 2008; 320(5876): 674-7.
[264]
Gordon S, Pl ddemann A. Macrophage Clearance of Apoptotic Cells: A Critical Assessment. Front Immunol 2018; 9: 127.
[265]
Obeid M, Tesniere A, Ghiringhelli F, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med 2007; 13(1): 54-61.
[266]
VanPatten S, Al-Abed Y. High Mobility Group Box-1 (HMGb1): Current Wisdom and Advancement as a Potential Drug Target. J Med Chem 2018; 61(12): 5093-107.
[267]
Andersson U, Wang H, Palmblad K, et al. High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes. J Exp Med 2000; 192(4): 565-70.
[268]
Manfredi AA, Capobianco A, Bianchi ME, Rovere-Querini P. Regulation of dendritic- and T-cell fate by injury-associated endogenous signals. Crit Rev Immunol 2009; 29(1): 69-86.
[269]
Wu T, Zhang W, Yang G, et al. HMGB1 overexpression as a prognostic factor for survival in cancer: A meta-analysis and systematic review. Oncotarget 2016; 7(31): 50417-27.
[270]
Shimada K, Crother TR, Karlin J, et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 2012; 36(3): 401-14.
[271]
Zhang Q, Raoof M, Chen Y, et al. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 2010; 464(7285): 104-7.
[272]
Vandenberk L, Garg AD, Verschuere T, et al. Irradiation of necrotic cancer cells, employed for pulsing dendritic cells (DCs), potentiates DC vaccine-induced antitumor immunity against high-grade glioma. OncoImmunology 2015; 5(2): e1083669.
[273]
Knoops B, Argyropoulou V, Becker S, Fert L, Kuznetsova O. Multiple Roles of Peroxiredoxins in Inflammation. Mol Cells 2016; 39(1): 60-4.
[274]
Linke B, Abeler-DArner L, Jahndel V, et al. The tolerogenic function of annexins on apoptotic cells is mediated by the annexin core domain. J Immunol 2015; 194(11): 5233-42.
[275]
Yoon KW, Byun S, Kwon E, et al. Control of signaling-mediated clearance of apoptotic cells by the tumor suppressor p53. Science 2015; 349(6247): 1261669.
[276]
Weyd H, Abeler-DArner L, Linke B, et al. Annexin A1 on the surface of early apoptotic cells suppresses CD8+ T cell immunity. PLoS One 2013; 8(4): e62449.
[277]
Green DR, Ferguson T, Zitvogel L, Kroemer G. Immunogenic and tolerogenic cell death. Nat Rev Immunol 2009; 9(5): 353-63.
[278]
Maeda A, Schwarz A, Kernebeck K, et al. Intravenous infusion of syngeneic apoptotic cells by photopheresis induces antigen-specific regulatory T cells. J Immunol 2005; 174(10): 5968-76.
[279]
Kaczmarek A, Vandenabeele P, Krysko DV. Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity 2013; 38(2): 209-23.
[280]
Kearney CJ, Cullen SP, Tynan GA, et al. Necroptosis suppresses inflammation via termination of TNF- or LPS-induced cytokine and chemokine production. Cell Death Differ 2015; 22(8): 1313-27.
[281]
Gong YN, Guy C, Olauson H, Becker JU, Yang M, Fitzgerald P, et al. ESCRT-III Acts Downstream of MLKL to Regulate Necroptotic Cell Death and Its Consequences 2017; 169(2): 286-300 e16.
[282]
Kepp O, Menger L, Vacchelli E, et al. Crosstalk between ER stress and immunogenic cell death. Cytokine Growth Factor Rev 2013; 24(4): 311-8.
[283]
Weiner LM, Lotze MT. Tumor-cell death, autophagy, and immunity. N Engl J Med 2012; 366(12): 1156-8.
[284]
Garg AD, De Ruysscher D, Agostinis P. Immunological metagene signatures derived from immunogenic cancer cell death associate with improved survival of patients with lung, breast or ovarian malignancies: A large-scale meta-analysis. OncoImmunology 2015; 5(2): e1069938.
[285]
Michaud M, Martins I, Sukkurwala AQ, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 2011; 334(6062): 1573-7.

© 2024 Bentham Science Publishers | Privacy Policy