Chonnam Med J.  2017 May;53(2):83-94. 10.4068/cmj.2017.53.2.83.

NIRF Heptamethine Cyanine Dye Nanocomplexes for Multi Modal Theranosis of Tumors

Affiliations
  • 1Department of Radiology, Chonnam National University Hwasun Hospital, Molecular Theranostics Laboratory, Hwasun, Korea. yjeong@jnu.ac.kr

Abstract

Heptamethine cyanine dyes are categorized as a class of near infrared fluorescent (NIRF) dyes which have been discovered to have tumor targeting and accumulation capability. This unique feature of NIRF dye makes it a promising candidate for imaging, targeted therapy and also as a drug delivery vehicle for various types of cancers. The favored uptake of dyes only in cancer cells is facilitated by several factors which include organic anion-transporting polypeptides, high mitochondrial membrane potential and tumor hypoxia in cancer cells. Currently nanotechnology has opened possibilities for multimodal or multifunctional strategies for cancer treatment. Including heptamethine cyanine dyes in nanoparticle based delivery systems have generally improved its theranostic ability by several fold owing to the multiple functionalities and structural features of heptamethine dyes. For this reason, nanocomplexes with NIRF heptamethine cyanine dye probe are preferred over non-targeting dyes such as indo cyanine green (ICG). This review sums up current trends and progress in NIRF heptamethine cyanine dye, including dye properties, multifunctional imaging and therapeutic applications in cancer.

Keyword

Neoplasms; Fluorescent Dyes; Drug Delivery Systems; Nanotechnology

MeSH Terms

Anoxia
Coloring Agents
Drug Delivery Systems
Fluorescent Dyes
Membrane Potential, Mitochondrial
Nanoparticles
Nanotechnology
Peptides
Theranostic Nanomedicine
Coloring Agents
Fluorescent Dyes
Peptides

Figure

  • FIG. 1 Schema showing several applications of heptamethine cyanine dyes.

  • FIG. 2 Different NIRF heptamethine dye structures. Figure reproduced with permission from (25) and (26). Copyright © 2017 Impact Journals and MDPI AG.

  • FIG. 3 Preferential uptake and retention of MHI-148. (A) MHI-148 chemical structure. (B) Ex vivo NIR fluorescence imaging showed increased MHI-148 dye uptake by different types of canine spontaneous tumors (blue arrows) as compared to adjacent normal tissues (white arrows). (C) Preferential uptake of NIRF dye in gastric cancer tissues relative to that in gastric tissues. In vivo NIRF imaging of mice bearing either orthotopic luc-tagged gastric tumor xenografts (left) or gastric ulcers (right). (D) NIRF imaging of clinical gastric cancer tissues. Schematic outlining the experimental procedures for NIRF imaging of freshly resected clinical gastric tumor tissues (left). Gross morphology and NIRF imaging of representative tumor tissues surgically resected from one of three gastric cancer patients. Representative images are presented in all panels. Original magnification: 4×; scale bars represent 4 mm. Figure images and the accompanying legend are reproduced with permission from (2530). Copyright © 2017 Impact Journals.

  • FIG. 4 IR780-liposome and IR780-phospholipid micelle developed for NIRF optical imaging. (A) Structure of IR780 iodide free dye, an IR780-liposome, and IR780-phospholipid micelle. (B) Real-time NIRF imaging of IR780-phospholipid micelles using the glioma spontaneous mouse model. Bioluminescence imaging (BLI) indicates the location and status of the U87MG ectopic tumor. Figure images and accompanying legend are reproduced with permission from (35). Copyright © 2017 American Chemical Society.

  • FIG. 5 Synthesis, in vivo MRI, and photothermal therapy of (A) Mn-IR825@PDA-PEG and (B) IR825@PAH-IONP-PEG. Figure images and accompanying legend are reproduced with permission from (3741). Copyright © 2012 American Chemical Society and John Wiley & Sons, Inc.

  • FIG. 6 Photothermal therapy of cancer cells (A) Laser-induced thermal damage to cancer cells after accumulation of NIRF probes. (B) Synthesis of IR780 loaded PMDPC-IR780 micelle nanoparticles and its application in photothermal therapy. Figure images and accompanying legend are reproduced with permission from (4953). Copyright © 2017 Dove Press Ltd and American Chemical Society.

  • FIG. 7 Photodynamic therapy by light-induced ROS release and damage to cancer cells after NIRF probe accumulation. Figure images and accompanying legend are reproduced with permission from (53). Copyright © 2017 Dove Press Ltd.

  • FIG. 8 Sonodynamic therapy using IR780 (A) Schema showing the action of IR780 in releasing ROS using a sonodynamic transducer. Upon receiving US, IR780 that had accumulated in the tumor cells would receive US energy. When in the excited-state, IR780 restored back to the ground-state and releasing energy; 1O2 and H2O absorb the released energy and changed into 1O2 and H2O2. The superfluous 1O2 and H2O2 would subsequently cause the apoptosis and necrosis of tumor cells. (B) Quantification of ROS release by the DCF-DA assay in 4T1 cancer cells for 1O2 (a), H2O2 (b), and ·OH (c). (C) Cell viability analysis of 4T1 breast cancer cells incubated with PBS, 4µM, 10µM, or 16µM of IR780. 4T1 breast cancer cells were incubated with PBS or IR780 and then administered with US for 0 s, 20 s, or 40 s. Twenty-four hours later, the levels of 1O2 were evaluated. (D) Photograph of 4T1 tumors removed from mice 30 days after the tumor-bearing mice were treated by SDT with IR780. Figure images and accompanying legend are reproduced from (60). Copyright © 2017 Macmillan Publishers Limited.

  • FIG. 9 DITSL nanoparticles for PTT/Chemotherapy. (A) Schematic diagram of DOX release from DITSL under NIR-laser irradiation. The liposome membrane temperature would increase when the NIR-laser irradiation was applied. Destruction of the liposome membrane occurs when the membrane temperature reaches 42℃. (B) Schematic diagram of DITSL preparation. (C) The representative infrared photothermal images of tumors following laser irradiation. Figure images and accompanying legend are reproduced from (63). Copyright © 2017 Ivyspring International Publisher.

  • FIG. 10 Schematic of Tf-IR780 nanoparticle preparation, in vitro phototherapy, and biodistribution mediated by Tf-IR780 NPs. (A) Transferrin self-assembly with IR-780 with the help of dithiothreitol (DTT) to form Tf-IR780 NPs (B) Fluorescence images of CT-26 cells expressing singlet oxygen indicated by H2DCFDA staining for detection under photoirradiation (1 W/cm2; 808 nm) for 5 min (Scale bar=20µm). (C) In vivo fluorescence imaging in mice bearing CT-26 tumors administered with Tf-IR780 NPs (0.3 mg/kg, IR780). Figure images and accompanying legend are reproduced from (68). Copyright © 2017 Macmillan Publishers Limited.


Reference

1. Jian WH, Yu TW, Chen CJ, Huang WC, Chiu HC, Chiang WH. Indocyanine green-encapsulated hybrid polymeric nanomicelles for photothermal cancer therapy. Langmuir. 2015; 31:6202–6210.
Article
2. Ma Y, Tong S, Bao G, Gao C, Dai Z. Indocyanine green loaded SPIO nanoparticles with phospholipid-PEG coating for dual-modal imaging and photothermal therapy. Biomaterials. 2013; 34:7706–7714.
Article
3. Miki K, Inoue T, Kobayashi Y, Nakano K, Matsuoka H, Yamauchi F, et al. Near-infrared dye-conjugated amphiphilic hyaluronic acid derivatives as a dual contrast agent for in vivo optical and photoacoustic tumor imaging. Biomacromolecules. 2015; 16:219–227.
Article
4. Wang H, Agarwal P, Zhao S, Yu J, Lu X, He X. A biomimetic hybrid nanoplatform for encapsulation and precisely controlled delivery of theranostic agents. Nat Commun. 2015; 6:10081.
Article
5. Yuan A, Wu J, Tang X, Zhao L, Xu F, Hu Y. Application of near-infrared dyes for tumor imaging, photothermal, and photodynamic therapies. J Pharm Sci. 2013; 102:6–28.
Article
6. Zhao P, Zheng M, Luo Z, Gong P, Gao G, Sheng Z, et al. NIR-driven smart theranostic nanomedicine for on-demand drug release and synergistic antitumour therapy. Sci Rep. 2015; 5:14258.
Article
7. Zhao P, Zheng M, Yue C, Luo Z, Gong P, Gao G, et al. Improving drug accumulation and photothermal efficacy in tumor depending on size of ICG loaded lipid-polymer nanoparticles. Biomaterials. 2014; 35:6037–6046.
Article
8. Zheng X, Xing D, Zhou F, Wu B, Chen WR. Indocyanine green-containing nanostructure as near infrared dual-functional targeting probes for optical imaging and photothermal therapy. Mol Pharm. 2011; 8:447–456.
Article
9. Espinosa A, Di Corato R, Kolosnjaj-Tabi J, Flaud P, Pellegrino T, Wilhelm C. Duality of iron oxide nanoparticles in cancer therapy: amplification of heating efficiency by magnetic hyperthermia and photothermal bimodal treatment. ACS Nano. 2016; 10:2436–2446.
Article
10. GhavamiNejad A, SamariKhalaj M, Aguilar LE, Park CH, Kim CS. pH/NIR light-controlled multidrug release via a mussel-inspired nanocomposite hydrogel for chemo-photothermal cancer therapy. Sci Rep. 2016; 6:33594.
Article
11. Jung HS, Han J, Lee JH, Lee JH, Choi JM, Kweon HS, et al. Enhanced NIR radiation-triggered hyperthermia by mitochondrial targeting. J Am Chem Soc. 2015; 137:3017–3023.
Article
12. Kim J, Oh J, Kang HW, Feldman MD, Milner TE. Photothermal response of superparamagnetic iron oxide nanoparticles. Lasers Surg Med. 2008; 40:415–421.
Article
13. Wang T, Wang D, Yu H, Wang M, Liu J, Feng B, et al. Intracellularly acid-switchable multifunctional micelles for combinational photo/chemotherapy of the drug-resistant tumor. ACS Nano. 2016; 10:3496–3508.
Article
14. Wang X, Zhang J, Wang Y, Wang C, Xiao J, Zhang Q, et al. Multi-responsive photothermal-chemotherapy with drug-loaded melanin-like nanoparticles for synergetic tumor ablation. Biomaterials. 2016; 81:114–124.
Article
15. Wang Z, Huang P, Jacobson O, Wang Z, Liu Y, Lin L, et al. Biomineralization-Inspired Synthesis of Copper Sulfide-Ferritin Nanocages as Cancer Theranostics. ACS Nano. 2016; 10:3453–3460.
Article
16. Wu M, Zhang D, Zeng Y, Wu L, Liu X, Liu J. Nanocluster of superparamagnetic iron oxide nanoparticles coated with poly (dopamine) for magnetic field-targeting, highly sensitive MRI and photothermal cancer therapy. Nanotechnology. 2015; 26:115102.
Article
17. Zhang N, Xu X, Zhang X, Qu D, Xue L, Mo R, et al. Nanocomposite hydrogel incorporating gold nanorods and paclitaxel-loaded chitosan micelles for combination photothermal-chemotherapy. Int J Pharm. 2016; 497:210–221.
Article
18. Zhou F, Wu S, Wu B, Chen WR, Xing D. Mitochondria-targeting single-walled carbon nanotubes for cancer photothermal therapy. Small. 2011; 7:2727–2735.
Article
19. Zhou Z, Sun Y, Shen J, Wei J, Yu C, Kong B, et al. Iron/iron oxide core/shell nanoparticles for magnetic targeting MRI and near-infrared photothermal therapy. Biomaterials. 2014; 35:7470–7478.
Article
20. Shi C, Wu JB, Pan D. Review on near-infrared heptamethine cyanine dyes as theranostic agents for tumor imaging, targeting, and photodynamic therapy. J Biomed Opt. 2016; 21:50901.
Article
21. Yang X, Shao C, Wang R, Chu CY, Hu P, Master V, et al. Optical imaging of kidney cancer with novel near infrared heptamethine carbocyanine fluorescent dyes. J Urol. 2013; 189:702–710.
Article
22. Yang X, Shi C, Tong R, Qian W, Zhau HE, Wang R, et al. Near IR heptamethine cyanine dye-mediated cancer imaging. Clin Cancer Res. 2010; 16:2833–2844.
Article
23. Luo S, Zhang E, Su Y, Cheng T, Shi C. A review of NIR dyes in cancer targeting and imaging. Biomaterials. 2011; 32:7127–7138.
Article
24. Yuan J, Yi X, Yan F, Wang F, Qin W, Wu G, et al. Near infrared fluorescence imaging of prostate cancer using heptamethine carbocyanine dyes. Mol Med Rep. 2015; 11:821–828.
Article
25. Shi C, Wu JB, Chu GC, Li Q, Wang R, Zhang C, et al. Heptamethine carbocyanine dye-mediated near-infrared imaging of canine and human cancers through the HIF-1α/OATPs signaling axis. Oncotarget. 2014; 5:10114–10126.
Article
26. Conceicao DS, Ferreira DP, Ferreira LF. Photochemistry and cytotoxicity evaluation of heptamethinecyanine Near Infrared (NIR) dyes. Int J Mol Sci. 2013; 14:18557–18571.
Article
27. Zhang C, Liu T, Su Y, Luo S, Zhu Y, Tan X, et al. A near-infrared fluorescent heptamethine indocyanine dye with preferential tumor accumulation for in vivo imaging. Biomaterials. 2010; 31:6612–6617.
Article
28. Wang Y, Liu T, Zhang E, Luo S, Tan X, Shi C. Preferential accumulation of the near infrared heptamethine dye IR-780 in the mitochondria of drug-resistant lung cancer cells. Biomaterials. 2014; 35:4116–4124.
Article
29. Wu JB, Shao C, Li X, Shi C, Li Q, Hu P, et al. Near-infrared fluorescence imaging of cancer mediated by tumor hypoxia and HIF1α/OATPs signaling axis. Biomaterials. 2014; 35:8175–8185.
Article
30. Zhao N, Zhang C, Zhao Y, Bai B, An J, Zhang H, et al. Optical imaging of gastric cancer with near-infrared heptamethine carbocyanine fluorescence dyes. Oncotarget. 2016; 7:57277–57289.
Article
31. Zhang E, Luo S, Tan X, Shi C. Mechanistic study of IR-780 dye as a potential tumor targeting and drug delivery agent. Biomaterials. 2014; 35:771–778.
Article
32. Buxhofer-Ausch V, Secky L, Wlcek K, Svoboda M, Kounnis V, Briasoulis E, et al. Tumor-specific expression of organic anion-transporting polypeptides: transporters as novel targets for cancer therapy. J Drug Deliv. 2013; 2013:863539.
33. Singh AK, Hahn MA, Gutwein LG, Rule MC, Knapik JA, Moudgil BM, et al. Multi-dye theranostic nanoparticle platform for bioimaging and cancer therapy. Int J Nanomedicine. 2012; 7:2739–2750.
34. Corem-Salkmon E, Perlstein B, Margel S. Design of near-infrared fluorescent bioactive conjugated functional iron oxide nanoparticles for optical detection of colon cancer. Int J Nanomedicine. 2012; 7:5517–5527.
35. Li S, Johnson J, Peck A, Xie Q. Near infrared fluorescent imaging of brain tumor with IR780 dye incorporated phospholipid nanoparticles. J Transl Med. 2017; 15:18.
Article
36. Lu C, Das S, Magut PK, Li M, El-Zahab B, Warner IM. Irradiation induced fluorescence enhancement in PEGylated cyanine-based NIR nano- and mesoscale GUMBOS. Langmuir. 2012; 28:14415–14423.
Article
37. Yang Y, Liu J, Liang C, Feng L, Fu T, Dong Z, et al. Nanoscale metal-organic particles with rapid clearance for magnetic resonance imaging-guided photothermal therapy. ACS Nano. 2016; 10:2774–2781.
Article
38. Yeh CS, Su CH, Ho WY, Huang CC, Chang JC, Chien YH, et al. Tumor targeting and MR imaging with lipophilic cyanine-mediated near-infrared responsive porous Gd silicate nanoparticles. Biomaterials. 2013; 34:5677–5688.
Article
39. Chen Q, Liang C, Wang X, He J, Li Y, Liu Z. An albumin-based theranostic nano-agent for dual-modal imaging guided photothermal therapy to inhibit lymphatic metastasis of cancer post surgery. Biomaterials. 2014; 35:9355–9362.
Article
40. Xiao L, Zhang Y, Yue W, Xie X, Wang JP, Chordia MD, et al. Heptamethine cyanine based (64)Cu-PET probe PC-1001 for cancer imaging: synthesis and in vivo evaluation. Nucl Med Biol. 2013; 40:351–360.
Article
41. Song X, Gong H, Liu T, Cheng L, Wang C, Sun X, et al. J-aggregates of organic dye molecules complexed with iron oxide nanoparticles for imaging-guided photothermal therapy under 915-nm light. Small. 2014; 10:4362–4370.
Article
42. Weber J, Beard PC, Bohndiek SE. Contrast agents for molecular photoacoustic imaging. Nat Methods. 2016; 13:639–650.
Article
43. Shi S, Liu Y, Chen Y, Zhang Z, Ding Y, Wu Z, et al. Versatile pH-response micelles with high cell-penetrating helical diblock copolymers for photoacoustic imaging guided synergistic chemo-photothermal therapy. Theranostics. 2016; 6:2170–2182.
Article
44. Okuda T, Kobayashi Y, Yanamoto S, Okamoto H. PEG conjugation of a near-infrared fluorescent probe for noninvasive dual imaging of lung deposition and gene expression by pulmonary gene delivery. J Drug Target. 2012; 20:801–812.
Article
45. Costantini P, Jacotot E, Decaudin D, Kroemer G. Mitochondrion as a novel target of anticancer chemotherapy. J Natl Cancer Inst. 2000; 92:1042–1053.
Article
46. Fulda S, Galluzzi L, Kroemer G. Targeting mitochondria for cancer therapy. Nat Rev Drug Discov. 2010; 9:447–464.
Article
47. Zhang E, Zhang C, Su Y, Cheng T, Shi C. Newly developed strategies for multifunctional mitochondria-targeted agents in cancer therapy. Drug Discov Today. 2011; 16:140–146.
Article
48. Wu JB, Shi C, Chu GC, Xu Q, Zhang Y, Li Q, et al. Near-infrared fluorescence heptamethine carbocyanine dyes mediate imaging and targeted drug delivery for human brain tumor. Biomaterials. 2015; 67:1–10.
Article
49. Chen Y, Li Z, Wang H, Wang Y, Han H, Jin Q, et al. IR-780 loaded phospholipid mimicking homopolymeric micelles for near-IR imaging and photothermal therapy of pancreatic cancer. ACS Appl Mater Interfaces. 2016; 8:6852–6858.
Article
50. Lin T, Yuan A, Zhao X, Lian H, Zhuang J, Chen W, et al. Self-assembled tumor-targeting hyaluronic acid nanoparticles for photothermal ablation in orthotopic bladder cancer. Acta Biomater. 2017; DOI: 10.1016/j.actbio.2017.02.021. [Epub ahead of print].
Article
51. Cheng L, He W, Gong H, Wang C, Chen Q, Cheng Z, et al. PEGylated micelle nanoparticles encapsulating a non-fluorescent near-infrared organic dye as a safe and highly-effective photothermal agent for in vivo cancer therapy. Adv Funct Mater. 2013; 23:5893–5902.
Article
52. Yuan A, Qiu X, Tang X, Liu W, Wu J, Hu Y. Self-assembled PEG-IR-780-C13 micelle as a targeting, safe and highly-effective photothermal agent for in vivo imaging and cancer therapy. Biomaterials. 2015; 51:184–193.
Article
53. Yi X, Wang F, Qin W, Yang X, Yuan J. Near-infrared fluorescent probes in cancer imaging and therapy: an emerging field. Int J Nanomedicine. 2014; 9:1347–1365.
Article
54. 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; 61:250–281.
Article
55. Felsher DW. Cancer revoked: oncogenes as therapeutic targets. Nat Rev Cancer. 2003; 3:375–380.
Article
56. Tan X, Luo S, Wang D, Su Y, Cheng T, Shi C. A NIR heptamethine dye with intrinsic cancer targeting, imaging and photosensitizing properties. Biomaterials. 2012; 33:2230–2239.
Article
57. Luo S, Tan X, Qi Q, Guo Q, Ran X, Zhang L, et al. A multifunctional heptamethine near-infrared dye for cancer theranosis. Biomaterials. 2013; 34:2244–2251.
Article
58. Pais-Silva C, de Melo-Diogo D, Correia IJ. IR780-loaded TPGSTOS micelles for breast cancer photodynamic therapy. Eur J Pharm Biopharm. 2017; 113:108–117.
Article
59. Wan GY, Liu Y, Chen BW, Liu YY, Wang YS, Zhang N. Recent advances of sonodynamic therapy in cancer treatment. Cancer Biol Med. 2016; 13:325–338.
Article
60. Li Y, Zhou Q, Deng Z, Pan M, Liu X, Wu J, et al. IR-780 dye as a sonosensitizer for sonodynamic therapy of breast tumor. Sci Rep. 2016; 6:25968.
Article
61. Zheng M, Yue C, Ma Y, Gong P, Zhao P, Zheng C, et al. Single-step assembly of DOX/ICG loaded lipid--polymer nanoparticles for highly effective chemo-photothermal combination therapy. ACS Nano. 2013; 7:2056–2067.
Article
62. Guo F, Yu M, Wang J, Tan F, Li N. Smart IR780 theranostic nanocarrier for tumor-specific therapy: hyperthermia-mediated bubble-generating and folate-targeted liposomes. ACS Appl Mater Interfaces. 2015; 7:20556–20567.
Article
63. Yan F, Duan W, Li Y, Wu H, Zhou Y, Pan M, et al. NIR-laser-controlled drug release from DOX/IR-780-loaded temperature-sensitive-liposomes for chemo-photothermal synergistic tumor therapy. Theranostics. 2016; 6:2337–2351.
Article
64. Peng CL, Chen YI, Liu HJ, Lee PC, Luo TY, Shieh MJ. A novel temperature-responsive micelle for enhancing combination therapy. Int J Nanomedicine. 2016; 11:3357–3369.
65. 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. 2016; 6:11070–11076.
Article
66. Jiang C, Cheng H, Yuan A, Tang X, Wu J, Hu Y. Hydrophobic IR780 encapsulated in biodegradable human serum albumin nanoparticles for photothermal and photodynamic therapy. Acta Biomater. 2015; 14:61–69.
Article
67. Luo S, Tan X, Fang S, Wang Y, Liu T, Wang X, et al. Mitochondria-targeted small-molecule fluorophores for dual modal cancer phototherapy. Adv Funct Mater. 2016; 26:2826–2835.
Article
68. Wang K, Zhang Y, Wang J, Yuan A, Sun M, Wu J, et al. Self-assembled IR780-loaded transferrin nanoparticles as an imaging, targeting and PDT/PTT agent for cancer therapy. Sci Rep. 2016; 6:27421.
Article
Full Text Links
  • CMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr