リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Photodynamic Therapy for Cancer using Mitochondrial Drug Delivery System」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Photodynamic Therapy for Cancer using Mitochondrial Drug Delivery System

Satrialdi 北海道大学

2020.03.25

概要

A non-invasive and specific targeting for cancer therapy is a necessity to manifest in order to minimize the harmful effect on the non-malignant cells. During the past century, photodynamic therapy (PDT) has been actively developed as a non-invasive approach to effectively eradicate the cancer cells with minimal effect on healthy cells. The PDT effect is derived from an energy transfer reaction between light as a source of energy to molecular oxygen, mediated by a light-activated molecule, known as the photosensitizer. The dynamic interaction among these three major components in PDT produces a lethal level of reactive oxygen species (ROS), mainly singlet oxygen. The selectivity of this therapy could be achieved by the specific accumulation of the non-toxic photosensitizer in the tumor region, accompanied by the precise delivery of light in the corresponding area. The singlet oxygen has a highly reactive characteristic that can readily react with several vital molecules in the biological system, resulting in the molecule dysfunction. This interaction may also lead to irreversible oxidative damage and further provoke a lethal effect for the cells. However, the harmful effects of singlet oxygen are restricted by their short lifetime and inadequate diffusion capacity. Therefore, the specific delivery of photosensitizer, mainly in organelle-level, could be a promising strategy to obtain the maximum benefits of this therapy. Moreover, as the important organelle that holds both the vital and lethal functions, mitochondria are identified to be an attractive target for optimizing the PDT outcomes.

In the current condition, the existing photosensitizers often manifest disadvantageous features for a practical PDT application. One of them is a non-specific accumulation either in the cellular or at the subcellular level. The other drawback is the inadequate ability of most photosensitizers in absorbing near-infrared (NIR) light. The application of NIR light in PDT, especially in the optical window of biological tissues, is profoundly beneficial because NIR light has an excellent penetration ability toward tissue consisting of water and biomolecules. These problems restrict the use of such compounds in clinical applications. Therefore, the development of a novel PDT system that can fulfill the requirements of the selective organelle accumulation in combination with the long-wavelength light activation is inevitable.

The main objective of this research was to construct a novel mitochondrial targeting PDT system with the long-wavelength light activation process. To realize that goal, a synergistic combination between a π-extended porphyrin-type photosensitizer, namely rTPA, and a MITO-Porter system, a versatile mitochondrial targeting liposomal-based nanodevice, was introduced. The incorporation of the rTPA compound into the MITO-Porter system was accomplished using the hydration method with the resulting particle showed a homogenous distribution with a diameter of 157 ± 7 nm and highly positive zeta potentials of 32 ± 3 mV. This novel mitochondrial targeting PDT system, namely the rTPA-MITO-Porter, manifested a robust capacity in producing a high level of singlet oxygen, specifically in the mitochondrial compartment of tumors, during a 700-nm light irradiation process. Based on the cellular uptake and intracellular observation results, most of the rTPA-MITO-Porter particles were efficiently internalized into the cells and concentrated in the mitochondrial compartment of tumors. Furthermore, this system displayed an efficient cytotoxicity profile against two types of human tumor cell lines, namely HeLa cells (human cervical cancer cells) and SAS cells (human squamous cells carcinoma of the tongue), as indicated by the low EC50 value of 0.16 ± 0.02 μM and 0.41 ± 0.18 μM, respectively. Additionally, the apoptosis pathway was actively induced during the PDT process of the rTPA-MITO-Porter, as shown by the formation of apoptotic bodies and fragmentation of mitochondrial structure.

Inspired by the excellent cell killing ability during the in vitro experiments, the translation process into the in vivo applications has further proceeded. A slight modification on the rTPA- MITO-Porter formulation, particularly on the helper lipid composition and the total lipids’ concentration, was made without altering the mitochondrial targeting ability and the photo- induced cytotoxic capacity. The remarkable inhibition of the tumor growth was obtained in the SAS cells-bearing mouse model after a single PDT treatment of the rTPA-MITO-Porter with the rTPA dose of 8.2 μg/mouse via intratumoral administration. There was also no significant alteration on the bodyweight of the mice during the treatment, implying the promising in vivo cell-killing ability of this system with a high safety profile. Furthermore, the depolarization on the mitochondrial membrane was observed after the PDT process of the rTPA-MITO-Porter, indicating the damage of mitochondrial membrane due to the specific localization of the photochemical reaction on the mitochondrial compartment of tumors. Finally, the findings presented in this research serve to verify the considerable functions of the MITO-Porter system as the mitochondrial selective drug delivery technology in potentiating the PDT outcomes as well as the importance of mitochondria as the predominant subcellular target for PDT. Moreover, this novel biologically-active nanomaterial manifests an encouraging feature for PDT applications, particularly for the superficial-type cancer cells.

この論文で使われている画像

参考文献

1. Finkel T. Signal transduction by reactive oxygen species. J Cell Biol. 194. 7-15 (2011).

2. Wiseman H, Halliwell B. Damage to DNA by reactive oxygen and nitrogen species: Role in inflammatory disease and progression to cancer. Biochem J. 313. 17-29 (1996).

3. Trachootham D, Alexandre J, Huang P. Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach?. Nat Rev Drug Discov. 8. 579–591 (2009).

4. Weinberg F, Hamanaka R, Wheaton WW, Weinberg S, Joseph J, Lopez M, Kalyanaraman B, Mutlu GM, Budinger GR, Chandel NS. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc Natl Acad Sci U S A. 107. 8788-8793 (2010).

5. Gamcsik MP, Kasibhatla MS, Teeter SD, Colvin OM. Glutathione levels in human tumors. Biomarkers. 17. 671-691 (2012).

6. Karlenius TC, Tonissen KF. Thioredoxin and cancer: A role for thioredoxin in all states of tumor oxygenation. Cancers. 2. 209-232 (2010).

7. Yoo MH, Xu XM, Carlson BA, Gladyshev VN, Hatfield DL. Thioredoxin reductase 1 deficiency reverses tumor phenotype and tumorigenicity of lung carcinoma cells. J Biol Chem. 281. 13005-13008 (2006).

8. Hawk MA, McCallister C, Schafer ZT. Antioxidant activity during tumor progression: A necessity for the survival of cancer cells?. Cancers. 8. 92 (2016).

9. Harris IS, Treloar AE, Inoue S, Sasaki M, Gorrini C, Lee KC, et al. Glutathione and Thioredoxin Antioxidant Pathways Synergize to Drive Cancer Initiation and Progression. Cancer Cell. 27. 211-222 (2015).

10. Piskounova E, Agathocleous M, Murphy MM, Hu Z, Huddlestun SE, Zhao Z, et al. Oxidative stress inhibits distant metastasis by human melanoma cells. Nature. 527. 186-191 (2015).

11. Gal K Le, Ibrahim MX, Wiel C, Sayin VI, Akula MK, Karlsson C, et al. Antioxidants can increase melanoma metastasis in mice. Sci Transl Med. 7. 308re8 (2015).

12. Sayin VI, Ibrahim MX, Larsson E, Nilsson JA, Lindahl P, Bergo MO. Cancer: Antioxidants accelerate lung cancer progression in mice. Sci Transl Med. 6. 221ra15 (2014).

13. Trachootham D, Zhou Y, Zhang H, Demizu Y, Chen Z, Pelicano H, et al. Selective killing of oncogenically transformed cells through a ROS-mediated mechanism by β-phenylethyl isothiocyanate. Cancer Cell. 10. 241-252 (2006).

14. Wang J, Luo B, Li X, Lu W, Yang J, Hu Y, Huang P, Wen S. Inhibition of cancer growth in vitro and in vivo by a novel ROS-modulating agent with ability to eliminate stem-like cancer cells. Cell Death Dis. 8. e2887 (2017).

15. Pelicano H, Feng L, Zhou Y, Carew JS, Hileman EO, Plunkett W, Keating MJ, Huang P. Inhibition of mitochondrial respiration: A novel strategy to enhance drug- induced apoptosis in human leukemia cells by a reactive oxygen species-mediated mechanism. J Biol Chem. 278. 37832-37839 (2003).

16. Dougherty TJ, E KJ, Goldfarb A, Weishaupt KR, Boyle D, Mittleman A. Photodynamic therapy for the treatment of malignant tumors. Can Res. 38. 2628-2635 (1978).

17. Dolmans DEJGJ, Fukumura D, Jain RK. Photodynamic therapy for cancer. Nat Rev Cancer. 3. 380-387 (2003).

18. Daniell MD, Hill JS. A history of photodynamic therapy. Aust N Z J Surg. 61. 340- 348 (1991).

19. Castano AP, Demidova TN, Hamblin M. Mechanisms in photodynamic therapy: part one. Photodiagnosis Photodyn Ther. 1. 279–293 (2004).

20. van Straten D, Mashayekhi V, de Bruijn HS, Oliveira S, Robinson DJ. Oncologic photodynamic therapy: Basic principles, current clinical status and future directions. Cancers. 9. 19 (2017).

21. Baskaran R, Lee J, Yang S-G. Clinical development of photodynamic agents and therapeutic applications. Biomater Res. 22. 25 (2018).

22. Abrahamse H, Hamblin MR. New photossensitizersfot photodynamic therapy. Biochem J. 473. 347-364 (2017).

23. Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, et al. Photodynamic Terapy of cancer: un update. CA Cancer J Clin. 61. 250-281 (2012).

24. Dysart JS, Patterson MS. Characterization of Photofrin photobleaching for singlet oxygen dose estimation during photodynamic therapy of MLL cells in vitro. Phys Med Biol. 50. 2597-2616 (2005).

25. Detty MR, Gibson SL, Wagner SJ. Current Clinical and Preclinical Photosensitizers for Use in Photodynamic Therapy. J Med Chem. 47. 3897-3915 (2004).

26. O’Connor AE, Gallagher WM, Byrne AT. Porphyrin and nonporphyrin photosensitizers in oncology: Preclinical and clinical advances in photodynamic therapy. Photochem Photobiol. 85. 1053-1074 (2009).

27. Brand MD, Affourtit C, Esteves TC, Green K, Lambert AJ, Miwa S, et al. Mitochondrial superoxide: Production, biological effects, and activation of uncoupling proteins. Free Radic Biol Med. 37. 755-767 (2004).

28. Kurokawa H, Ito H, Inoue M, Tabata K, Sato Y, Yamagata K, et al. High resolution imaging of intracellular oxygen concentration by phosphorescence lifetime. Sci Rep. 5. 10657 (2015).

29. Guo F, Yu M, Wang J, Tan F, Li N. The mitochondria-targeted and IR780-regulated theranosomes for imaging and enhanced photodynamic/photothermal therapy. RSC Adv. 6. 11070-11076 (2016).

30. Mahalingam SM, Ordaz JD, Low PS. Targeting of a Photosensitizer to the Mitochondrion Enhances the Potency of Photodynamic Therapy. ACS Omega. 3. 6066-6074 (2018).

31. Lv W, Zhang Z, Zhang KY, Yang H, Liu S, Xu A, et al. A Mitochondria-Targeted Photosensitizer Showing Improved Photodynamic Therapy Effects Under Hypoxia. Angew Chemie - Int Ed. 55. 9947-9951 (2016).

32. Murphy MP. Selective targeting of bioactive compounds to mitochondria. Trends Biotechnol. 15. 326-330 (1997).

33. Yamada Y, Akita H, Kamiya H, Kogure K, Yamamoto T, Shinohara Y, Yamashita K, Kobayashi H, Kikuchi H, Harashima H. MITO-Porter: A liposome-based carrier system for delivery of macromolecules into mitochondria via membrane fusion. Biochim et Biophys Acta. 1778. 423-432 (2008).

34. Yamada Y, Harashima H. Mitochondrial drug delivery systems for macromolecule and their therapeutic application to mitochondrial diseases. Adv Drug Deliv Rev. 60. 1439-1462 (2008).

35. Khalil IA, Kogure K, Futaki S, Harashima H. High density of octaarginine stimulates macropinocytosis leading to efficient intracellular trafficking for gene expression. J Biol Chem. 281. 3544-3551 (2006).

36. Yamada Y, Fukuda Y, Harashima H. An analysis of membrane fusion between mitochondrial double membranes and MITO-Porter, mitochondrial fusogenic vesicles. Mitochondrion. 24. 50-55 (2015).

37. Takano Y, Munechika R, Biju V, Harashima H, Imahori H, Yamada Y. Optical control of mitochondrial reductive reactions in living cells using an electron donor- acceptor linked molecule. Nanoscale. 9. 18690-18698 (2017).

38. Sternberg ED, Dolphin D, Brückner C. Porphyrin-based photosensitizers for use in photodynamic therapy. Tetrahedron. 54. 4151-4202 (1998).

39. Takano Y, Numata T, Fujishima K, Miyake K, Nakao K, Grove WD, et al. Optical control of neuronal firing: Via photoinduced electron transfer in donor-acceptor conjugates. Chem Sci. 7. 3331-3337 (2016).

40. Andreoni A, Cubeddu R, De Silvestri S, Laporta P, Jori G, Reddi E. Hematoporphyrin derivative: experimental evidence for aggregated species. Chem Phys Lett. 88. 33-36 (1982).

41. Moreira LM, Dos Santos FV, Lyon JP, Maftoum-Costa M, Pacheco-Soares C, Soares Da Silva N. Photodynamic therapy: Porphyrins and phthalocyanines as photosensitizers. Aust J Chem. 61. 741-754 (2008).

42. TAnielian C, HEINRICH G. Effect of aggregation on the hematoporphyrin‐ sensitized production of singlet molecular oxygen. Photochem Photobiol. 61. 131- 135 (1995).

43. Bangham AD, Horne RW. Negative staining of phospholipids and their structural modification by surface-active agents as observed in the electron microscope. J Mol Biol. 8. 660-668 (1964).

44. Papahadjopoulos D. Liposomes as Drug Carriers. Annu Rep Med Chem. 14. 250-260 (1979).

45. Akbarzadeh A, Rezaei-Sadabady R, Davaran S, Joo SW, Zarghami N, Hanifehpour Y, et al. Liposome: Classification, preparation, and applications. Nanoscale Res Lett. 8. 102 (2013).

46. Bozzuto G, Molinari A. Liposomes as nanomedical devices. Int J Nanomedicine. 10. 975-999 (2015).

47. Kim S, Fujitsuka M, Majima T. Photochemistry of singlet oxygen sensor green. J Phys Chem B. 117. 13985-13992 (2013).

48. Kim S, Tachikawa T, Fujitsuka M, Majima T. Far-red fluorescence probe for monitoring singlet oxygen during photodynamic therapy. J Am Chem Soc. 136. 11707-11715 (2014).

49. Zorov DB, Filburn CR, Klotz LO, Zweier JL, Sollott SJ. Reactive oxygen species (ROS)-induced ROS release: a new phenomenon accompanying induction of the mitochondrial permeability transition in cardiac myocytes. J Exp Med. 192. 1001- 1014 (2000).

50. Yamada Y, Nakamura K, Furukawa R, Kawamura E, Moriwaki T, Matsumoto K, et al. Mitochondrial delivery of bongkrekic acid using a MITO-porter prevents the induction of apoptosis in human hela cells. J Pharm Sci. 102. 1008-1015 (2013).

51. Yamada Y, Hashida M, Harashima H. Hyaluronic acid controls the uptake pathway and intracellular trafficking of an octaarginine-modified gene vector in CD44 positive- and CD44 negative-cells. Biomaterials. 52. 189-198 (2015).

52. Schindelin J, Arganda-carreras I, Frise E, Kaynig V, Pietzsch T, Preibisch S, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods. 9. 676-682 (2012).

53. Adler J, Parmryd I. Quantifying colocalization by correlation: The pearson correlation coefficient is superior to the Mander’s overlap coefficient. Cytom Part A. 77. 733-742 (2010).

54. Chennoufi R, Bougherara H, Gagey-Eilstein N, Dumat B, Henry E, Subra F, et al. Mitochondria-targeted Triphenylamine Derivatives Activatable by Two-Photon Excitation for Triggering and Imaging Cell Apoptosis. Sci Rep. 6. 21458 (2016).

55. Soriano J, Mora-Espí I, Alea-Reyes ME, Pérez-García L, Barrios L, Ibáñez E, et al. Cell death mechanisms in Tumoral and Non-Tumoral human cell lines triggered by photodynamic treatments: Apoptosis, necrosis and parthanatos. Sci Rep. 7. 41340 (2017).

56. Kessel D, Oleinick NL. Chapter 1 Initiation of Autophagy by Photodynamic Therapy. Methods Enzymol. 453. 1-16 (2009).

57. Mroz P, Yaroslavsky A, Kharkwal GB, Hamblin MR. Cell death pathways in photodynamic therapy of cancer. Cancers. 3. 2516-2539 (2011).

58. Mahalingam SM, Ordaz JD, Low PS. Targeting of a Photosensitizer to the Mitochondrion Enhances the Potency of Photodynamic Therapy. ACS Omega. 3. 6066-6074 (2018).

59. Mitsunaga M, Ogawa M, Kosaka N, Rosenblum LT, Choyke PL, Kobayashi H. Cancer cell-selective in vivo near infrared photoimmunotherapy targeting specific membrane molecules. Nat Med. 17. 1685-1691 (2011).

60. JF Kerr, AH Wyllie, AR Currie. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 26. 239-257 (1972).

61. Taylor RC, Cullen SP, Martin SJ. Apoptosis: Controlled demolition at the cellular level. Nat Rev Mol Cell Biol. 9. 231-241 (2008).

62. Qi T, Chen B, Wang Z, Du H, Liu D, Yin Q, et al. A pH-Activatable nanoparticle for dual-stage precisely mitochondria-targeted photodynamic anticancer therapy. Biomaterials. 213. 119219 (2019).

63. Yu Z, Sun Q, Pan W, Li N, Tang B. A Near-Infrared Triggered Nanophotosensitizer Inducing Domino Effect on Mitochondrial Reactive Oxygen Species Burst for Cancer Therapy. ACS Nano. 9. 11064-11074 (2015).

64. Futaki S, Nakase I. Cell-Surface Interactions on Arginine-Rich Cell-Penetrating Peptides Allow for Multiplex Modes of Internalization. Acc Chem Res. 50. 2449- 2456 (2017).

65. El-Sayed A, Khalil IA, Kogure K, Futaki S, Harashima H. Octaarginine- and octalysine-modified nanoparticles have different modes of endosomal escape. J Biol Chem. 283. 23450-23461 (2008).

66. Farhood H, Serbina N, Huang L. The role of dioleoyl phosphatidylethanolamine in cationic liposome mediated gene transfer. Biochim Biophys Acta. 1235. 289-295 (1995).

67. Ricchelli F, Gobbo S, Moreno G, Salet C, Brancaleon L, Mazzini A. Photophysical properties of porphyrin planar aggregates in liposomes. Eur J Biochem. 253. 760- 765 (1998).

68. Kirakci K, Zelenka J, Rumlová M, Cvačka J, Ruml T, Lang K. Cationic octahedral molybdenum cluster complexes functionalized with mitochondria-targeting ligands: Photodynamic anticancer and antibacterial activities. Biomater Sci. 7. 1386-1392 (2019).

69. Tang PM, Liu XZ, Zhang DM, Fong WP, Fung KP. Pheophorbide a based photodynamic therapy induces apoptosis via mitochondrial-mediated pathway in human uterine carcinosarcoma. Cancer Biol Ther. 8. 533-539 (2009).

70. Zhou Z, Liu J, Rees TW, Wang H, Li X, Chao H, et al. Heterometallic Ru–Pt metallacycle for two-photon photodynamic therapy. Proc Natl Acad Sci. 115. 5664- 5669 (2018).

71. Pereira PMR, Silva S, Bispo M, Zuzarte M, Gomes C, Girão H, et al. Mitochondria- Targeted Photodynamic Therapy with a Galactodendritic Chlorin to Enhance Cell Death in Resistant Bladder Cancer Cells. Bioconjug Chem. 27. 2762-2769 (2016).

72. Thomas AP, Palanikumar L, Jeena MT, Kim K, Ryu JH. Cancer-mitochondria- targeted photodynamic therapy with supramolecular assembly of HA and a water soluble NIR cyanine dye. Chem Sci. 8. 8351-8356 (2017).

73. Zhang D, Wen L, Huang R, Wang H, Hu X, Xing D. Mitochondrial specific photodynamic therapy by rare-earth nanoparticles mediated near-infrared graphene quantum dots. Biomaterials. 153. 14-26 (2018).

74. Taratula O, Schumann C, Naleway MA, Pang AJ, Chon KJ, Taratula O. A multifunctional theranostic platform based on phthalocyanine-loaded dendrimer for image-guided drug delivery and photodynamic therapy. Mol Pharm. 10. 3946- 3958 (2013).

75. Machacek M, Demuth J, Cermak P, Vavreckova M, Hruba L, Jedlickova A, et al. Tetra(3,4-pyrido)porphyrazines Caught in the Cationic Cage: Toward Nanomolar Active Photosensitizers. J Med Chem. 59. 9443-9456 (2016).

76. Vachova L, Machacek M, Kučera R, Demuth J, Cermak P, Kopecky K, et al. Heteroatom-substituted tetra(3,4- pyrido)porphyrazines: A stride toward near- infrared-absorbing macrocycles. Org Biomol Chem. 13. 5608-5612 (2015).

77. Liu J, Chen Y, Li G, Zhang P, Jin C, Zeng L, et al. Ruthenium(II) polypyridyl complexes as mitochondria-targeted two-photon photodynamic anticancer agents. Biomaterials. 56. 140-153 (2015).

78. Li X, Zheng BY, Ke MR, Zhang Y, Huang JD, Yoon J. A tumor-pH-responsive supramolecular photosensitizer for activatable photodynamic therapy with minimal in vivo skin phototoxicity. Theranostics. 7. 2746-2756 (2017).

79. Pan D, Zhong X, Zhao W, Yu Z, Yang Z, Wang D, et al. Meso-substituted porphyrin photosensitizers with enhanced near-infrared absorption: Synthesis, characterization and biological evaluation for photodynamic therapy. Tetrahedron. 74. 2677-2683 (2018).

80. Lei Z, Zhang X, Zheng X, Liu S, Xie Z. Porphyrin-ferrocene conjugates for photodynamic and chemodynamic therapy. Org Biomol Chem. 16. 8613-8619 (2018).

81. Liu DZ, Chen WY, Tasi LM, Yang SP. Microcalorimetric and shear studies on the effects of cholesterol on the physical stability of lipid vesicles. Colloids Surf A Physicochem Eng Asp. 172. 57-67 (2000).

82. Briuglia ML, Rotella C, McFarlane A, Lamprou DA. Influence of cholesterol on liposome stability and on in vitro drug release. Drug Deliv Transl Res. 5. 231-242 (2015).

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る