AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (3.5 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review | Open Access

Targeting-specific Nanoprobes in the Second Near-infrared Window for Biomedical Applications

Yutong Guo1Zhaoyue Li1Bingchen Guo1Bo Wang1Yingfeng Tu1( )
Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
Show Author Information

Graphical Abstract

Abstract

Fluorescence imaging with high spatiotemporal resolution and sensitivity is employed for in vivo visualization and detection applications. Compared with visible light and the first near-infrared window (700–900 nm), the second near-infrared window (1 000–1 700 nm) imaging has a lower auto-background fluorescence, deeper tissue penetration, and a higher signal-to-noise ratio, thus highlighting its broad prospects in the biomedical field. Currently, reported second near-infrared region probes include single-walled carbon nanotubes, rare-earth nanoparticles, quantum dots, metal materials, and organic molecular dyes. Multimodal imaging can overcome the limitations of single imaging and provide more comprehensive information on the anatomical structure, tissue composition, and cellular and molecular quantification of lesions to achieve multi-level visualization. Therefore, second near-infrared window nanoprobes must be engineered for multimodal imaging. Moreover, these nanoprobes can be actively targeted by modification with antibodies, peptides, nucleotides, or biofilms to obtain detailed and accurate information on biological systems. This review describes the active targeting capabilities of various second near-infrared window nanoprobes in multimodal imaging, diagnosis, and treatment of different diseases by carrying different ligands and the common features and future application prospects of second near-infrared window fluorescent nanoprobes with targeting ability.

References

[1]

G.D. Luker, K.E. Luker. Optical imaging: Current applications and future directions. Journal of Nuclear Medicine, 2008, 49(1): 1−4. https://doi.org/10.2967/jnumed.107.045799

[2]

J. Cao, B. Zhu, K. Zheng, et al. Recent progress in NIR-II contrast agent for biological imaging. Front Bioeng Biotechnol, 2019, 7: 487. https://doi.org/10.3389/fbioe.2019.00487

[3]

N.N. Zhang, C.Y. Lu, M.J. Chen, et al. Recent advances in near-infrared II imaging technology for biological detection. Journal of Nanobiotechnology, 2021, 19(1): 132. https://doi.org/10.1186/s12951-021-00870-z

[4]

S.A. Hilderbrand, R. Weissleder. Near-infrared fluorescence: application to in vivo molecular imaging. Current Opinion in Chemical Biology, 2010, 14(1): 71−79. https://doi.org/10.1016/j.cbpa.2009.09.029

[5]

S. Luo, E. Zhang, Y. Su, et al. A review of NIR dyes in cancer targeting and imaging. Biomaterials, 2011, 32(29): 7127−7138. https://doi.org/10.1016/j.biomaterials.2011.06.024

[6]

J. Zhao, D. Zhong, S. Zhou. NIR-I-to-NIR-II fluorescent nanomaterials for biomedical imaging and cancer therapy. Journal of Materials Chemistry B, 2018, 6(3): 349−365. https://doi.org/10.1039/C7TB02573D

[7]

S.Q. He, J. Song, J.L. Qu, et al. Crucial breakthrough of second near-infrared biological window fluorophores: Design and synthesis toward multimodal imaging and theranostics. Chemical Society Reviews, 2018, 47(12): 4258−4278. https://doi.org/10.1039/c8cs00234g

[8]

Kenry, Y.K. Duan, B. Liu. Recent advances of optical imaging in the second near-infrared window. Advanced Materials, 2018, 30(47): 1802394. https://doi.org/10.1002/adma.201802394

[9]

K. Welsher, Z. Liu, S.P. Sherlock, et al. A route to brightly fluorescent carbon nanotubes for near-infrared imaging in mice. Nature Nanotechnology, 2009, 4(11): 773−780. https://doi.org/10.1038/nnano.2009.294

[10]

Y. Fan, F. Zhang. A new generation of NIR-II probes: Lanthanide-based nanocrystals for bioimaging and biosensing. Advanced Optical Materials, 2019, 7(7): 1801417. https://doi.org/10.1002/adom.201801417

[11]

Y.J. Zhang, H.C. Yang, X.Y. An, et al. Controlled synthesis of Ag2Te@Ag2S core–shell quantum dots with enhanced and tunable fluorescence in the second near-infrared window. Small, 2020, 16(14): e2001003. https://doi.org/10.1002/smll.202001003

[12]

H. Liu, G. Hong, Z. Luo, et al. Atomic-precision gold clusters for NIR-II imaging. Advanced Materials, 2019, 31(46): e1901015. https://doi.org/10.1002/adma.201901015

[13]

Q. Li, C.J. Zeman, Z. Ma, et al. Bright NIR-II photoluminescence in rod-shaped icosahedral gold nanoclusters. Small, 2021, 17(11): e2007992. https://doi.org/10.1002/smll.202007992

[14]

W.Z. Wang, Z.R. Ma, S.J. Zhu, et al. Molecular cancer imaging in the second near-infrared window using a renal-excreted NIR-II fluorophore-peptide probe. Advanced Materials, 2018, 30(22): 1800106. https://doi.org/10.1002/adma.201800106

[15]

Y. Li, D. Hu, Z. Sheng, et al. Self-assembled AIEgen nanoparticles for multiscale NIR-II vascular imaging. Biomaterials, 2021, 264: 120365. https://doi.org/10.1016/j.biomaterials.2020.120365

[16]

S.Y. Lee, S.I. Jeon, S. Jung, et al. Targeted multimodal imaging modalities. Advanced Drug Delivery Reviews, 2014, 76: 60−78. https://doi.org/10.1016/j.addr.2014.07.009

[17]

A. Alaarg, C. Pérez-Medina, J.M. Metselaar, et al. Applying nanomedicine in maladaptive inflammation and angiogenesis. Advanced Drug Delivery Reviews, 2017, 119: 143−158. https://doi.org/10.1016/j.addr.2017.05.009

[18]

S.J. Zhu, R. Tian, A.L. Antaris, et al. Near-infrared-II molecular dyes for cancer imaging and surgery. Advanced Materials, 2019, 31(24): 1900321. https://doi.org/10.1002/adma.201900321

[19]

J.C. Shen, J. Karges, K. Xiong, et al. Cancer cell membrane camouflaged iridium complexes functionalized black-titanium nanoparticles for hierarchical-targeted synergistic NIR-II photothermal and sonodynamic therapy. Biomaterials, 2021, 275: 120979. https://doi.org/10.1016/j.biomaterials.2021.120979

[20]

N. Bertrand, J. Wu, X.Y. Xu, et al. Cancer nanotechnology: The impact of passive and active targeting in the era of modern cancer biology. Advanced Drug Delivery Reviews, 2014, 66: 2−25. https://doi.org/10.1016/j.addr.2013.11.009

[21]

R.P. Das, V.V. Gandhi, B.G. Singh, et al. Passive and active drug targeting: Role of nanocarriers in rational design of anticancer formulations. Current Pharmaceutical Design, 2019, 25(28): 3034−3056. https://doi.org/10.2174/1381612825666190830155319

[22]

N. Kamaly, Z. Xiao, P.M. Valencia, et al. Targeted polymeric therapeutic nanoparticles: Design, development and clinical translation. Chemical Society Reviews, 2012, 41(7): 2971−3010. https://doi.org/10.1039/c2cs15344k

[23]

H. Gong, R. Peng, Z. Liu. Carbon nanotubes for biomedical imaging: The recent advances. Advanced Drug Delivery Reviews, 2013, 65(15): 1951−1963. https://doi.org/10.1016/j.addr.2013.10.002

[24]
J.T. Robinson, G.S. Hong, Y.Y. Liang, et al. In vivo fluorescence imaging in the second near-infrared window with long circulating carbon nanotubes capable of ultrahigh tumor uptake. Journal of the American Chemical Society, 2012, 134(25): 10664–10669.
[25]

Y. Yomogida, T. Tanaka, M.F. Zhang, et al. Industrial-scale separation of high-purity single-chirality single-wall carbon nanotubes for biological imaging. Nature Communications, 2016, 7: 12056. https://doi.org/10.1038/ncomms12056

[26]

H. Yi, D. Ghosh, M.H. Ham, et al. M13 phage-functionalized single-walled carbon nanotubes As nanoprobes for second near-infrared window fluorescence imaging of targeted tumors. Nano Letters, 2012, 12(3): 1176−1183. https://doi.org/10.1021/nl2031663

[27]

L. Ceppi, N.M. Bardhan, Y. Na, et al. Real-time single-walled carbon nanotube-based fluorescence imaging improves survival after debulking surgery in an ovarian cancer model. ACS Nano, 2019, 13(5): 5356−5365. https://doi.org/10.1021/acsnano.8b09829

[28]

C. Kofoed Andersen, S. Khatri, J. Hansen, et al. Carbon nanotubes-potent carriers for targeted drug delivery in rheumatoid arthritis. Pharmaceutics, 2021, 13(4): 453. https://doi.org/10.3390/pharmaceutics13040453

[29]

Z. Yu, C. Eich, L.J. Cruz. Recent advances in rare-earth-doped nanoparticles for NIR-II imaging and cancer theranostics. Frontiers in Chemistry, 2020, 8: 496. https://doi.org/10.3389/fchem.2020.00496

[30]

H.X. Zhang, Z.H. Chen, X. Liu, et al. A mini-review on recent progress of new sensitizers for luminescence of lanthanide doped nanomaterials. Nano Research, 2020, 13(7): 1795−1809. https://doi.org/10.1007/s12274-020-2661-8

[31]

J. Zhou, Q. Liu, W. Feng, et al. Upconversion luminescent materials: Advances and applications. Chemical Reviews, 2015, 115(1): 395−465. https://doi.org/10.1021/cr400478f

[32]
Y.T. Zhong, Z.R. Ma, F.F. Wang, et al. In vivo molecular imaging for immunotherapy using ultra-bright near-infrared-IIb rare-earth nanoparticles. Nature Biotechnology, 2019, 37(11): 1322–1331.
[33]

N.J. Johnson, S. He, S. Diao, et al. Direct evidence for coupled surface and concentration quenching dynamics in lanthanide-doped nanocrystals. Journal of the American Chemical Society, 2017, 139(8): 3275−3282. https://doi.org/10.1021/jacs.7b00223

[34]

A. Dong, X. Ye, J. Chen, Y. Kang, et al. A generalized ligand-exchange strategy enabling sequential surface functionalization of colloidal nanocrystals. Journal of the American Chemical Society, 2011, 133(4): 998−1006. https://doi.org/10.1021/ja108948z

[35]

F. Danhier, A. Le Breton, V. Préat. RGD-based strategies to target alpha(v) beta(3) integrin in cancer therapy and diagnosis. Molecular Pharmaceutics, 2012, 9(11): 2961−2973. https://doi.org/10.1021/mp3002733

[36]

P.Y. Wang, X.D. Wang, Q. Luo, et al. Fabrication of red blood cell-based multimodal theranostic probes for second near-infrared window fluorescence imaging-guided tumor surgery and photodynamic therapy. Theranostics, 2019, 9(2): 369−380. https://doi.org/10.7150/thno.29817

[37]

B. Lin, Y. Wang, K. Zhao, et al. Exosome-based rare earth nanoparticles for targeted in situ and metastatic tumor imaging with chemo-assisted immunotherapy. Biomaterials Science, 2022, 10(3): 744−752. https://doi.org/10.1039/D1BM01809D

[38]

D. Ghosh, A.F. Bagley, Y.J. Na, et al. Deep, noninvasive imaging and surgical guidance of submillimeter tumors using targeted M13-stabilized single-walled carbon nanotubes. Proc Natl Acad Sci USA, 2014, 111(38): 13948−13953. https://doi.org/10.1073/pnas.1400821111

[39]

J.J. Zhang, Y. Lin, H. Zhou, et al. Cell membrane-camouflaged NIR II fluorescent Ag2 Te quantum dots-based nanobioprobes for enhanced in vivo homotypic tumor imaging. Advanced Healthcare Materials, 2019, 8(14): e1900341. https://doi.org/10.1002/adhm.201900341

[40]

H. Yang, R. Li, Y. Zhang, et al. Colloidal alloyed quantum dots with enhanced photoluminescence quantum yield in the NIR-II window. Journal of the American Chemical Society, 2021, 143(6): 2601−2607. https://doi.org/10.1021/jacs.0c13071

[41]
W.L. Li, G.L. Zhang, , Liu, L. Near-infrared inorganic nanomaterials for precise diagnosis and therapy. Frontiers in Bioengineering and Biotechnology, 2021, 9: 768927.
[42]
Y.P. Du, , Xu, B., Fu, T., Cai, M., Li, F., Zhang, Y., Wang, Q. B. Near-infrared photoluminescent Ag2S quantum dots from a single source precursor. Journal of the American Chemical Society, 2010, 132(5): 1470–1471.
[43]

B. Purushothaman, J.M. Song. Ag2S quantum dot theragnostics. Biomaterials Science, 2021, 9(1): 51−69. https://doi.org/10.1039/d0bm01576h

[44]

Q. Wu, L. Jiang, S.C. Li, et al. Small molecule inhibitors targeting the PD-1/PD-L1 signaling pathway. Acta Pharmacologica Sinica, 2021, 42(1): 1−9. https://doi.org/10.1038/s41401-020-0366-x

[45]

F.F. Wang, H. Wan, Z.R. Ma, et al. Light-sheet microscopy in the near-infrared II window. Nature Methods, 2019, 16(6): 545−552. https://doi.org/10.1038/s41592-019-0398-7

[46]

G.T. Yu, M.Y. Luo, H. Li, et al. Molecular targeting nanoprobes with non-overlap emission in the second near-infrared window for in vivo two-color colocalization of immune cells. ACS Nano, 2019, 13(11): 12830−12839. https://doi.org/10.1021/acsnano.9b05038

[47]

M. Wang, H. Li, B. Huang, et al. An ultra-stable, oxygen-supply nanoprobe emitting in near-infrared-II window to guide and enhance radiotherapy by promoting anti-tumor immunity. Advanced Healthcare Materials, 2021, 10(12): e2100090. https://doi.org/10.1002/adhm.202100090

[48]

S. Jeong, Y. Jung, S. Bok, et al. Multiplexed in vivo imaging using size-controlled quantum dots in the second near-infrared window. Advanced Healthcare Materials, 2018, 7(24): e1800695. https://doi.org/10.1002/adhm.201800695

[49]

P. Wang, Y. Fan, L. Lu, et al. NIR-II nanoprobes in-vivo assembly to improve image-guided surgery for metastatic ovarian cancer. Nature Communications, 2018, 9(1): 2898. https://doi.org/10.1038/s41467-018-05113-8

[50]

P.Y. Wang, S.H. Jiang, Y. Li, et al. Downshifting nanoprobes with follicle stimulating hormone peptide fabrication for highly efficient NIR II fluorescent bioimaging guided ovarian tumor surgery. Nanomedicine:Nanotechnology,Biology and Medicine, 2020, 28: 102198. https://doi.org/10.1016/j.nano.2020.102198

[51]

B. Lin, J. Wu, Y. Wang, et al. Peptide functionalized upconversion/NIR II luminescent nanoparticles for targeted imaging and therapy of oral squamous cell carcinoma. Biomaterials Science, 2021, 9(3): 1000−1007. https://doi.org/10.1039/d0bm01737j

[52]

R. Lv, Y. Wang, B. Lin, et al. Targeted luminescent probes for precise upconversion/NIR II luminescence diagnosis of lung adenocarcinoma. Analytical Chemistry, 2021, 93(11): 4984−4992. https://doi.org/10.1021/acs.analchem.1c00374

[53]

X. Song, S. Li, H. Guo, et al. Graphene-oxide-modified lanthanide nanoprobes for tumor-targeted visible/NIR-II luminescence imaging. Angewandte Chemie, 2019, 58(52): 18981−18986. https://doi.org/10.1002/anie.201909416

[54]

Z.M. Tao, X.N. Dang, X. Huang, et al. Early tumor detection afforded by in vivo imaging of near-infrared II fluorescence. Biomaterials, 2017, 134: 202−215. https://doi.org/10.1016/j.biomaterials.2017.04.046

[55]

B.T. Luk, L.F. Zhang. Cell membrane-camouflaged nanoparticles for drug delivery. Journal of Controlled Release, 2015, 220: 600−607. https://doi.org/10.1016/j.jconrel.2015.07.019

[56]

P. Moitra, M. Alafeef, K. Dighe, et al. Synthesis and characterisation of N-gene targeted NIR-II fluorescent probe for selective localisation of SARS-CoV-2. Chemical Communications, 2021, 57(51): 6229−6232. https://doi.org/10.1039/d1cc01410b

[57]

Z.N. Wu, Q.F. Yao, O.J.H. Chai, et al. Unraveling the impact of gold(I)–thiolate motifs on the aggregation-induced emission of gold nanoclusters. Angewandte Chemie, 2020, 132(25): 10020−10025. https://doi.org/10.1002/anie.201916675

[58]

Z. Miao, Z. Gao, R. Chen, et al. Surface-bioengineered gold nanoparticles for biomedical applications. Current Medicinal Chemistry, 2018, 25(16): 1920−1944. https://doi.org/10.2174/0929867325666180117111404

[59]

Y. Tao, M. Li, J. Ren, et al. Metal nanoclusters: Novel probes for diagnostic and therapeutic applications. Chemical Society Reviews, 2015, 44(23): 8636−8663. https://doi.org/10.1039/c5cs00607d

[60]

G. Huang, H. Huang. Hyaluronic acid-based biopharmaceutical delivery and tumor-targeted drug delivery system. Journal of Control Release, 2018, 278: 122−126. https://doi.org/10.1016/j.jconrel.2018.04.015

[61]

R.X. Cui, W. Sun, M.Y. Liu, et al. Near-infrared emissive lanthanide metal–organic frameworks for targeted biological imaging and pH-controlled chemotherapy. ACS Applied Materials &Interfaces, 2021, 13(49): 59164−59173. https://doi.org/10.1021/acsami.1c20817

[62]

H. Wu, P. Jia, Y. Zou, et al. Cascade targeting tumor mitochondria with CuS nanoparticles for enhanced photothermal therapy in the second near-infrared window. Biomaterials Science, 2021, 9(15): 5209−5217. https://doi.org/10.1039/d1bm00589h

[63]

Q. Wen, Y. Zhang, C. Li, et al. NIR-II fluorescent self-assembled peptide nanochain for ultrasensitive detection of peritoneal metastasis. Angewandte Chemie, 2019, 58(32): 11001−11006. https://doi.org/10.1002/anie.201905643

[64]

C.Y. Li, W.F. Li, H.H. Liu, et al. An activatable NIR-II nanoprobe for in vivo early real-time diagnosis of traumatic brain injury. Angewandte Chemie, 2020, 59(1): 247−252. https://doi.org/10.1002/anie.201911803

[65]

S. Mateos, J. Lifante, C. Li, et al. Instantaneous in vivo imaging of acute myocardial infarct by NIR-II luminescent nanodots. Small, 2020, 16(29): e1907171. https://doi.org/10.1002/smll.201907171

[66]

P. Zhu, S.S. Gao, H. Lin, et al. Inorganic nanoshell-stabilized liquid metal for targeted photonanomedicine in NIR-II biowindow. Nano Letters, 2019, 19(3): 2128−2137. https://doi.org/10.1021/acs.nanolett.9b00364

[67]

Y. Zhang, F. Lv, Y. Cheng, et al. Pd@Au bimetallic nanoplates decorated mesoporous MnO2 for synergistic nucleus-targeted NIR-II photothermal and hypoxia-relieved photodynamic therapy. Advanced Healthcare Materials, 2020, 9(2): e1901528. https://doi.org/10.1002/adhm.201901528

[68]

Z. Liu, K. Qiu, X. Liao, et al. Nucleus-targeting ultrasmall ruthenium(iv) oxide nanoparticles for photoacoustic imaging and low-temperature photothermal therapy in the NIR-II window. Chemical Communications, 2020, 56(20): 3019−3022. https://doi.org/10.1039/C9CC09728G

[69]

C. Wu, D. Wang, M. Cen, et al. Mitochondria-targeting NO gas nanogenerator for augmenting mild photothermal therapy in the NIR-II biowindow. Chemical Communications, 2020, 56(92): 14491−14494. https://doi.org/10.1039/d0cc05125j

[70]

P. Ruenraroengsak, D. Kiryushko, I.G. Theodorou, et al. Frizzled-7-targeted delivery of zinc oxide nanoparticles to drug-resistant breast cancer cells. Nanoscale, 2019, 11(27): 12858−12870. https://doi.org/10.1039/c9nr01277j

[71]

B. Li, Z.Y. Jiang, D.Y. Xie, et al. Cetuximab-modified CuS nanoparticles integrating near-infrared-II-responsive photothermal therapy and anti-vessel treatment. International Journal of Nanomedicine, 2018, 13: 7289−7302. https://doi.org/10.2147/ijn.S175334

[72]

H. Yang, H.P. He, Z.R. Tong, et al. The impact of size and surface ligand of gold nanorods on liver cancer accumulation and photothermal therapy in the second near-infrared window. Journal of Colloid and Interface Science, 2020, 565: 186−196. https://doi.org/10.1016/j.jcis.2020.01.026

[73]

H. Lu, F. Yang, B. Liu, et al. Intracellular low-abundance microRNA imaging by a NIR-assisted entropy-driven DNA system. Nanoscale Horiz, 2019, 4(2): 472−479. https://doi.org/10.1039/c8nh00330k

[74]

J.H. Zeng, M. Wu, S.Y. Lan, et al. Facile preparation of biocompatible Ti2O3 nanoparticles for second near-infrared window photothermal therapy. Journal of Materials Chemistry B, 2018, 6(47): 7889−7897. https://doi.org/10.1039/c8tb02079e

[75]

S.J. Zhu, B.C. Yung, S. Chandra, et al. Near-infrared-II (NIR-II) bioimaging via off-peak NIR-I fluorescence emission. Theranostics, 2018, 8(15): 4141−4151. https://doi.org/10.7150/thno.27995

[76]

C. Li, G. Chen, Y. Zhang, et al. Advanced fluorescence imaging technology in the near-infrared-II window for biomedical applications. Journal of the American Chemical Society, 2020, 142(35): 14789−14804. https://doi.org/10.1021/jacs.0c07022

[77]

H.L. Yu, M. Ji. Recent advances of organic near-infrared II fluorophores in optical properties and imaging functions. Molecular Imaging and Biology, 2021, 23(2): 160−172. https://doi.org/10.1007/s11307-020-01545-1

[78]

Y. Wu, W. Zhu. Organic sensitizers from D-π-A to D-A-π-A: Effect of the internal electron-withdrawing units on molecular absorption, energy levels and photovoltaic performances. Chemical Society Reviews, 2013, 42(5): 2039−2058. https://doi.org/10.1039/c2cs35346f

[79]

G. Qian, Z.Y. Wang. Design, synthesis, and properties of benzobisthiadiazole-based donor–π–acceptor–π–donor type of low-band-gap chromophores and polymers. Canadian Journal of Chemistry, 2010, 88(3): 192−201. https://doi.org/10.1139/v09-157

[80]

Y. Zhu, C. Gu, Y. Miao, et al. D-a polymers for fluorescence/photoacoustic imaging and characterization of their photothermal properties. Journal of Materials Chemistry B, 2019, 7(42): 6576−6584. https://doi.org/10.1039/c9tb01196j

[81]

Y. Wang, W. Zhang, P. Sun, et al. A novel multimodal NIR-II nanoprobe for the detection of metastatic lymph nodes and targeting chemo-photothermal therapy in oral squamous cell carcinoma. Theranostics, 2019, 9(2): 391−404. https://doi.org/10.7150/thno.30268

[82]

Y.N. Dai, Z.Q. Sun, H.H. Zhao, et al. NIR-II fluorescence imaging guided tumor-specific NIR-II photothermal therapy enhanced by starvation mediated thermal sensitization strategy. Biomaterials, 2021, 275: 120935. https://doi.org/10.1016/j.biomaterials.2021.120935

[83]

H. Zhang, D. Yu, S. Liu, et al. NIR-II hydrogen-bonded organic frameworks (HOFs) used for target-specific amyloid-β photooxygenation in an Alzheimer’s disease model. Angewandte Chemie, 2022, 61(2): e202109068. https://doi.org/10.1002/anie.202109068

[84]

R. Tian, H. Ma, Q. Yang, et al. Rational design of a super-contrast NIR-II fluorophore affords high-performance NIR-II molecular imaging guided microsurgery. Chemical Science, 2019, 10(1): 326−332. https://doi.org/10.1039/c8sc03751e

[85]

R. Tian, Q. Zeng, S. Zhu, et al. Albumin-chaperoned cyanine dye yields superbright NIR-II fluorophore with enhanced pharmacokinetics. Science Advances, 2019, 5(9): eaaw0672. https://doi.org/10.1126/sciadv.aaw0672

[86]

X.D. Zeng, Y.L. Xiao, J.C. Lin, et al. Near-infrared II dye-protein complex for biomedical imaging and imaging-guided photothermal therapy. Advanced Healthcare Materials, 2018, 7(18): e1800589. https://doi.org/10.1002/adhm.201800589

[87]

C. Yao, Y. Chen, M. Zhao, et al. A bright, renal-clearable NIR-II brush macromolecular probe with long blood circulation time for kidney disease bioimaging. Angewandte Chemie, 2022, 61(5): e202114273. https://doi.org/10.1002/anie.202114273

[88]

W.W. Wu, Y. Yang, Z.Y. Liang, et al. Near infrared II laser controlled free radical releasing nanogenerator for synergistic nitric oxide and alkyl radical therapy of breast cancer. Nanoscale, 2021, 13: 11169−11187. https://doi.org/10.1039/d1nr01859k

[89]

Y. Wu, C. Wang, J. Guo, et al. An RGD modified water-soluble fluorophore probe for in vivo NIR-II imaging of thrombosis. Biomaterials Science, 2020, 8(16): 4438−4446. https://doi.org/10.1039/d0bm00729c

[90]

Y. He, S. Wang, P. Yu, et al. NIR-II cell endocytosis-activated fluorescent probes for in vivo high-contrast bioimaging diagnostics. Chemical Science, 2021, 12(31): 10474−10482. https://doi.org/10.1039/d1sc02763h

[91]

M. Zhang, W. Wang, M. Mohammadniaei, et al. Upregulating aggregation-induced-emission nanoparticles with blood-tumor-barrier permeability for precise photothermal eradication of brain tumors and induction of local immune responses. Advanced Materials, 2021, 33(22): e2008802. https://doi.org/10.1002/adma.202008802

[92]

J.F. Wang, Y.S. Liu, M. Morsch, et al. Brain-targeted aggregation-induced-emission nanoparticles with near-infrared imaging at 1550nm boosts orthotopic glioblastoma theranostics. Advanced Materials, 2022, 34(5): 2106082. https://doi.org/10.1002/adma.202106082

[93]

H. Zhou, X. Zeng, A. Li, et al. Upconversion NIR-II fluorophores for mitochondria-targeted cancer imaging and photothermal therapy. Nature Communications, 2020, 11(1): 6183. https://doi.org/10.1038/s41467-020-19945-w

[94]

Y. Li, X. Hu, W. Yi, et al. NIR-II fluorescence imaging of skin avulsion and necrosis. Frontiers in Chemistry, 2019, 7: 696. https://doi.org/10.3389/fchem.2019.00696

[95]

W. Yi, H. Zhou, A. Li, et al. A NIR-II fluorescent probe for articular cartilage degeneration imaging and osteoarthritis detection. Biomaterials Science, 2019, 7(3): 1043−1051. https://doi.org/10.1039/c8bm01440j

[96]

S. Kurbegovic, K. Juhl, H. Chen, et al. Molecular targeted NIR-II probe for image-guided brain tumor surgery. Bioconjugate Chemistry, 2018, 29(11): 3833−3840. https://doi.org/10.1021/acs.bioconjchem.8b00669

[97]

Y. Sun, C. Qu, H. Chen, et al. Novel benzo-bis(1, 2, 5-thiadiazole) fluorophores for in vivo NIR-II imaging of cancer. Chemical Science, 2016, 7(9): 6203−6207. https://doi.org/10.1039/c6sc01561a

[98]

H. Zhou, W. Yi, A. Li, et al. Specific small-molecule NIR-II fluorescence imaging of osteosarcoma and lung metastasis. Advanced Healthcare Materials, 2020, 9(1): e1901224. https://doi.org/10.1002/adhm.201901224

[99]

X. Zhou, Q. Liu, W. Yuan, et al. Ultrabright NIR-II emissive polymer dots for metastatic ovarian cancer detection. Advanced Science, 2021, 8(4): 2000441. https://doi.org/10.1002/advs.202000441

[100]

A.L. Antaris, H. Chen, K. Cheng, et al. A small-molecule dye for NIR-II imaging. Nature Materials, 2016, 15(2): 235−242. https://doi.org/10.1038/nmat4476

[101]

H. Wan, H. Ma, S. Zhu, et al. Developing a bright NIR-II fluorophore with fast renal excretion and its application in molecular imaging of immune checkpoint PD-L1. Advanced Functional Materials, 2018, 28(50): 1804956. https://doi.org/10.1002/adfm.201804956

[102]

Q. Liu, J.W. Tian, Y. Tian, et al. Near-infrared-II nanoparticles for cancer imaging of immune checkpoint programmed death-ligand 1 and photodynamic/immune therapy. ACS Nano, 2021, 15(1): 515−525. https://doi.org/10.1021/acsnano.0c05317

[103]

X.P. Luo, D.H. Hu, D.Y. Gao, et al. Metabolizable near-infrared-II nanoprobes for dynamic imaging of deep-seated tumor-associated macrophages in pancreatic cancer. ACS Nano, 2021, 15(6): 10010−10024. https://doi.org/10.1021/acsnano.1c01608

[104]

J.A. Chen, H.M. Pan, Z.J. Wang, et al. Imaging of ovarian cancers using enzyme activatable probes with second near-infrared window emission. Chemical Communications, 2020, 56(18): 2731−2734. https://doi.org/10.1039/c9cc09158k

[105]

Y. Feng, S.J. Zhu, A.L. Antaris, et al. Live imaging of follicle stimulating hormone receptors in gonads and bones using near infrared II fluorophore. Chemical Science, 2017, 8(5): 3703−3711. https://doi.org/10.1039/c6sc04897h

[106]
C. Xu, Y.Y. Jiang, J.S. Huang, et al Second near-infrared light-activatable polymeric nanoantagonist for photothermal immunometabolic cancer therapy. Advanced Materials, 2021, 33(36): 2101410.
[107]
Y.F. Tang, Y.Y. Li, X.M. Hu, et al. “dual lock-and-key” -controlled nanoprobes for ultrahigh specific fluorescence imaging in the second near-infrared window. Advanced Materials, 2018, 30(31): 1801140.
[108]

L. He, F. Qing, M. Li, et al. Paclitaxel/IR1061-co-loaded protein nanoparticle for tumor-targeted and pH/NIR-II-triggered synergistic photothermal-chemotherapy. International Journal of Nanomedicine, 2020, 15: 2337−2349. https://doi.org/10.2147/ijn.S240707

[109]

Z.R. Ma, H. Wan, W.Z. Wang, et al. A theranostic agent for cancer therapy and imaging in the second near-infrared window. Nano Research, 2019, 12(2): 273−279. https://doi.org/10.1007/s12274-018-2210-x

[110]

K.H. Xue, H.N. Tian, F.K. Zhu, et al. Ultralong-circulating and self-targeting “watson–crick A = T” -inspired supramolecular nanotheranostics for NIR-II imaging-guided photochemotherapy. ACS Applied Materials &Interfaces, 2020, 12(29): 32477−32492. https://doi.org/10.1021/acsami.0c09090

[111]

Q. Wang, J.Z. Xu, R.Y. Geng, et al. High performance one-for-all phototheranostics: NIR-II fluorescence imaging guided mitochondria-targeting phototherapy with a single-dose injection and 808 nm laser irradiation. Biomaterials, 2020, 231: 119671. https://doi.org/10.1016/j.biomaterials.2019.119671

[112]

S.J. Liu, C. Chen, Y.Y. Li, et al. Constitutional isomerization enables bright NIR-II AIEgen for brain-inflammation imaging. Advanced Functional Materials, 2020, 30(7): 1908125. https://doi.org/10.1002/adfm.201908125

[113]

P.K. Upputuri, M. Pramanik. Photoacoustic imaging in the second near-infrared window: A review. Journal of Biomedical Optics, 2019, 24(4): 040901. https://doi.org/10.1117/1.Jbo.24.4.040901

[114]

S. Wang, J. Lin, T. Wang, et al. Recent advances in photoacoustic imaging for deep-tissue biomedical applications. Theranostics, 2016, 6(13): 2394−2413. https://doi.org/10.7150/thno.16715

[115]

Q. Chen, Z. Zheng, X. He, et al. A tumor-targeted theranostic nanomedicine with strong absorption in the NIR-II biowindow for image-guided multi-gradient therapy. Journal of Materials Chemistry B, 2020, 8(41): 9492−9501. https://doi.org/10.1039/d0tb01915a

[116]

Y. Zhang, Q. Shen, Q. Li, et al. Ultrathin two-dimensional plasmonic PtAg nanosheets for broadband phototheranostics in both NIR-I and NIR-II biowindows. Advanced Science, 2021, 8(17): e2100386. https://doi.org/10.1002/advs.202100386

[117]

X.L. Guo, C.C. Wen, Q.X. Xu, et al. A full-spectrum responsive B-TiO2@SiO2–HA nanotheranostic system for NIR-II photoacoustic imaging-guided cancer phototherapy. Journal of Materials Chemistry B, 2021, 9(8): 2042−2053. https://doi.org/10.1039/d0tb02952a

[118]
C. Wu, Y. Zhang, Z. Li, et al. A novel photoacoustic nanoprobe of ICG@PEG-Ag2S for atherosclerosis targeting and imaging in vivo. Nanoscale, 2016, 8(25): 12531–12539.
[119]

H. Karabeber, R. Huang, P. Iacono, et al. Guiding brain tumor resection using surface-enhanced Raman scattering nanoparticles and a hand-held Raman scanner. ACS Nano, 2014, 8(10): 9755−9766. https://doi.org/10.1021/nn503948b

[120]

J. Sun, J. Wang, W. Hu, et al. Camouflaged gold nanodendrites enable synergistic photodynamic therapy and NIR biowindow II photothermal therapy and multimodal imaging. ACS Applied Materials &Interfaces, 2021, 13(9): 10778−10795. https://doi.org/10.1021/acsami.1c01238

[121]

L. Zhang, S. Xue, F. Ren, et al. An atherosclerotic plaque-targeted single-chain antibody for MR/NIR-II imaging of atherosclerosis and anti-atherosclerosis therapy. Journal of Nanobiotechnology, 2021, 19(1): 296. https://doi.org/10.1186/s12951-021-01047-4

[122]
Y.J. Liu, H.H. Liu, H.X. Yan, et al. Aggregation-induced absorption enhancement for deep near-infrared II photoacoustic imaging of brain gliomas in vivo. Advanced Science, 2019, 6: 1801615.
[123]

M.Z. Xu, B. Xue, Y. Wang, et al. Temperature-feedback nanoplatform for NIR-II penta-modal imaging-guided synergistic photothermal therapy and CAR-NK immunotherapy of lung cancer. Small, 2021, 17(43): e2101397. https://doi.org/10.1002/smll.202101397

[124]

H. Yan, J. Chen, Y. Li, et al. Ultrasmall hybrid protein-copper sulfide nanoparticles for targeted photoacoustic imaging of orthotopic hepatocellular carcinoma with a high signal-to-noise ratio. Biomaterials Science, 2018, 7(1): 92−103. https://doi.org/10.1039/c8bm00767e

[125]

H.C. Zhou, J. Ren, Y. Lin, et al. Intravital NIR-II three-dimensional photoacoustic imaging of biomineralized copper sulfide nanoprobes. Journal of Materials Chemistry B, 2021, 9(13): 3005−3014. https://doi.org/10.1039/d0tb03010d

[126]

K. Cheng, H. Chen, Jenkins, C. H., et al. Synthesis, characterization, and biomedical applications of a targeted dual-modal near-infrared-II fluorescence and photoacoustic imaging nanoprobe. ACS Nano, 2017, 11(12): 12276−12291. https://doi.org/10.1021/acsnano.7b05966

[127]

J. Chen, J. Qi, C. Chen, et al. Tocilizumab-conjugated polymer nanoparticles for NIR-II photoacoustic-imaging-guided therapy of rheumatoid arthritis. Advanced Materials, 2020, 32(37): e2003399. https://doi.org/10.1002/adma.202003399

[128]

J. Wu, H.J. Lee, L.Y. You, et al. Functionalized NIR-II semiconducting polymer nanoparticles for single-cell to whole-organ imaging of PSMA-positive prostate cancer. Small, 2020, 16(19): e2001215. https://doi.org/10.1002/smll.202001215

[129]

Y.N. Dai, H.H. Zhao, K. He, et al. NIR-II excitation phototheranostic nanomedicine for fluorescence/photoacoustic tumor imaging and targeted photothermal-photonic thermodynamic therapy. Small, 2021, 17(42): e2102527. https://doi.org/10.1002/smll.202102527

[130]

Y. Li, J. Lin, P. Wang, et al. Tumor microenvironment responsive shape-reversal self-targeting virus-inspired nanodrug for imaging-guided near-infrared-II photothermal chemotherapy. ACS Nano, 2019, 13(11): 12912−12928. https://doi.org/10.1021/acsnano.9b05425

[131]

K. Xue, F. Wei, J. Lin, et al. Tumor acidity-responsive carrier-free nanodrugs based on targeting activation via ICG-templated assembly for NIR-II imaging-guided photothermal-chemotherapy. Biomaterials Science, 2021, 9(3): 1008−1019. https://doi.org/10.1039/d0bm01864c

[132]

C. Zhu, Z. Ding, Z. Guo, et al. Full-spectrum responsive ZrO2-based phototheranostic agent for NIR-II photoacoustic imaging-guided cancer phototherapy. Biomaterials Science, 2020, 8(23): 6515−6525. https://doi.org/10.1039/d0bm01482f

[133]

Z. Wei, F. Xue, F. Xin, et al. A thieno-isoindigo derivative-based conjugated polymer nanoparticle for photothermal therapy in the NIR-II bio-window. Nanoscale, 2020, 12(38): 19665−19672. https://doi.org/10.1039/d0nr03771k

[134]

X. Geng, D. Gao, D. Hu, et al. Active-targeting NIR-II phototheranostics in multiple tumor models using platelet-camouflaged nanoprobes. ACS Applied Materials &Interfaces, 2020, 12(50): 55624−55637. https://doi.org/10.1021/acsami.0c16872

[135]

D. Zhu, M. Lyu, Q. Huang , et al. Stellate plasmonic exosomes for penetrative targeting tumor NIR-II thermo-radiotherapy. ACS Applied Materials &Interfaces, 2020, 12(33): 36928−36937. https://doi.org/10.1021/acsami.0c09969

[136]

Y. Suo, F. Wu, P. Xu , et al. NIR-II fluorescence endoscopy for targeted imaging of colorectal cancer. Advanced Healthcare Materials, 2019, 8(23): e1900974. https://doi.org/10.1002/adhm.201900974

[137]

C. Song, F. Li, X. Guo , et al. Gold nanostars for cancer cell-targeted SERS-imaging and NIR light-triggered plasmonic photothermal therapy (PPTT) in the first and second biological windows. Journal of Materials Chemistry B, 2019, 7(12): 2001−2008. https://doi.org/10.1039/c9tb00061e

[138]

J. Wu, J. Liu, B. et al. Met-targeted dual-modal MRI/NIR II imaging for specific recognition of head and neck squamous cell carcinoma. ACS Biomaterials Science &Engineering, 2021, 7(4): 1640−1650. https://doi.org/10.1021/acsbiomaterials.0c01807

[139]

Y. Sun, X. Zeng, Y. Xiao , et al. Novel dual-function near-infrared II fluorescence and PET probe for tumor delineation and image-guided surgery. Chemical Science, 2018, 9(8): 2092−2097. https://doi.org/10.1039/c7sc04774f

[140]

D.S. Wei, Y.J. Yu, Y. Huang, et al. A near-infrared-II polymer with tandem fluorophores demonstrates superior biodegradability for simultaneous drug tracking and treatment efficacy feedback. ACS Nano, 2021, 15(3): 5428−5438. https://doi.org/10.1021/acsnano.1c00076

[141]

J. Wang, J. Sun, Y. Wang, et al. Gold nanoframeworks with mesopores for raman-photoacoustic imaging and photo-chemo tumor therapy in the second near-infrared biowindow. Advanced Functional Materials, 2020, 30(9): 1908825. https://doi.org/10.1002/adfm.201908825

[142]

S. Liang, M. Sun, Y. Lu , et al. Cytokine-induced killer cells-assisted tumor-targeting delivery of Her-2 monoclonal antibody-conjugated gold nanostars with NIR photosensitizer for enhanced therapy of cancer. Journal of Materials Chemistry B, 2020, 8(36): 8368−8382. https://doi.org/10.1039/d0tb01391a

[143]
M. Zhao, J. Ding, Q. Mao , et al. A novel αvβ3 integrin-targeted NIR-II nanoprobe for multimodal imaging-guided photothermal therapy of tumors in vivo. Nanoscale, 2020, 12(13): 6953–6958.
[144]

Y. Cao, T. Wu, K. Zhang , et al. Engineered exosome-mediated near-infrared-II region V2C quantum dot delivery for nucleus-target low-temperature photothermal therapy. ACS Nano, 2019, 13: 1499−1510. https://doi.org/10.1021/acsnano.8b07224

[145]

C. Zhang, W. Sun, Y. Wang , et al. Gd-/ CuS-loaded functional nanogels for MR/PA imaging-guided tumor-targeted photothermal therapy. ACS Applied Materials &Interfaces, 2020, 12(8): 9107−9117. https://doi.org/10.1021/acsami.9b23413

Nano Biomedicine and Engineering
Pages 135-151
Cite this article:
Guo Y, Li Z, Guo B, et al. Targeting-specific Nanoprobes in the Second Near-infrared Window for Biomedical Applications. Nano Biomedicine and Engineering, 2024, 16(2): 135-151. https://doi.org/10.26599/NBE.2024.9290061

797

Views

161

Downloads

1

Crossref

1

Scopus

Altmetrics

Received: 31 July 2023
Revised: 25 August 2023
Accepted: 24 October 2023
Published: 05 February 2024
© The Author(s) 2024.

This is an open-access article distributed under  the  terms  of  the  Creative  Commons  Attribution  4.0 International  License (CC BY) (http://creativecommons.org/licenses/by/4.0/), which  permits  unrestricted  use,  distribution,  and reproduction in any medium, provided the original author and source are credited.

Return