Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Targeted multicolor in vivo imaging over 1,000 nm enabled by nonamethine cyanines

Abstract

Recent progress has shown that using wavelengths between 1,000 and 2,000 nm, referred to as the shortwave-infrared or near-infrared (NIR)-II range, can enable high-resolution in vivo imaging at depths not possible with conventional optical wavelengths. However, few bioconjugatable probes of the type that have proven invaluable for multiplexed imaging in the visible and NIR range are available for imaging these wavelengths. Using rational design, we have generated persulfonated indocyanine dyes with absorbance maxima at 872 and 1,072 nm through catechol-ring and aryl-ring fusion, respectively, onto the nonamethine scaffold. Multiplexed two-color and three-color in vivo imaging using monoclonal antibody and dextran conjugates in several tumor models illustrate the benefits of concurrent labeling of the tumor and healthy surrounding tissue and lymphatics. These efforts are enabled by complementary advances in a custom-built NIR/shortwave-infrared imaging setup and software package for multicolor real-time imaging.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Synthesis and characterization of nonamethine cyanines.
Fig. 2: Probe comparison, multiplexing strategy and single-color monoclonal antibody imaging.
Fig. 3: Applications of in vivo multicolor optical imaging.

Similar content being viewed by others

Data availability

Image datasets, including all raw and processed imaging data generated in this work, are available at BioImage Archive (accession number S-BIAD325). Source data are provided with this paper. All other datasets are included in the Supplementary Information.

Code availability

Custom code (ccda-v3.0.1beta) used to collect data in this work is available at GitHub (https://gitlab.com/brunslab/ccda/).

References

  1. Nguyen, Q. T. & Tsien, R. Y. Fluorescence-guided surgery with live molecular navigation–a new cutting edge. Nat. Rev. Cancer 13, 653–662 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Salo, D., Zhang, H., Kim, D. M. & Berezin, M. Y. Multispectral measurement of contrast in tissue-mimicking phantoms in near-infrared spectral range of 650 to 1,600 nm. J. Biomed. Opt. 19, 086008 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Carr, J. A. et al. Absorption by water increases fluorescence image contrast of biological tissue in the shortwave infrared. Proc. Natl Acad. Sci. USA 115, 9080–9085 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Pittet, M. J. & Weissleder, R. Intravital imaging. Cell 147, 983–991 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Zhu, B., Kwon, S., Rasmussen, J. C., Litorja, M. & Sevick-Muraca, E. M. Comparison of NIR versus SWIR fluorescence image device performance using working standards calibrated with SI units. IEEE Trans. Med. Imaging 39, 944–951 (2020).

    Article  PubMed  Google Scholar 

  6. Thimsen, E., Sadtler, B. & Berezin, M. Y. Shortwave-infrared (SWIR) emitters for biological imaging: a review of challenges and opportunities. Nanophotonics 6, 1043–1054 (2017).

    Article  CAS  Google Scholar 

  7. Zhu, S., Tian, R., Antaris, A. L., Chen, X. & Dai, H. Near-infrared-II molecular dyes for cancer imaging and surgery. Adv. Mater. 31, e1900321 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Hong, G. S. et al. Multifunctional in vivo vascular imaging using near-infrared II fluorescence. Nat. Med. 18, 1841–1846 (2012).

    Article  PubMed Central  Google Scholar 

  9. Hong, G. S. et al. Through-skull fluorescence imaging of the brain in a new near-infrared window. Nat. Photonics 8, 723–730 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Franke, D. et al. Continuous injection synthesis of indium arsenide quantum dots emissive in the short-wavelength infrared. Nat. Commun. 7, 12749 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Bruns, O. T. et al. Next-generation in vivo optical imaging with short-wave infrared quantum dots. Nat. Biomed. Eng. https://doi.org/10.1038/s41551-017-0056 (2017).

  12. Liu, Q. et al. Sub-10 nm hexagonal lanthanide-doped NaLuF4 upconversion nanocrystals for sensitive bioimaging in vivo. J. Am. Chem. Soc. 133, 17122–17125 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Chen, Y. et al. Shortwave infrared in vivo imaging with gold nanoclusters. Nano Lett. 17, 6330–6334 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Starosolski, Z. et al. Indocyanine green fluorescence in second near-infrared (NIR-II) window. PLoS ONE 12, e0187563 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Carr, J. A. et al. Shortwave infrared fluorescence imaging with the clinically approved near-infrared dye indocyanine green. Proc. Natl Acad. Sci. USA 115, 4465–4470 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Antaris, A. L. et al. A small-molecule dye for NIR-II imaging. Nat. Mater. https://doi.org/10.1038/nmat4476 (2015).

  17. Wan, H. et al. A bright organic NIR-II nanofluorophore for three-dimensional imaging into biological tissues. Nat. Commun. 9, 1171 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Godard, A. et al. Water-soluble Aza-BODIPYs: biocompatible organic dyes for high contrast in vivo NIR-II imaging. Bioconjug. Chem. 31, 1088–1092 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Swamy, M. M. M., Murai, Y., Monde, K., Tsuboi, S. & Jin, T. Shortwave-infrared fluorescent molecular imaging probes based on pi-conjugation extended indocyanine green. Bioconjug. Chem. 32, 1541–1547 (2021).

    Article  CAS  PubMed  Google Scholar 

  20. Waggoner, A. Fluorescent labels for proteomics and genomics. Curr. Opin. Chem. Biol. 10, 62–66 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Levitus, M. & Ranjit, S. Cyanine dyes in biophysical research: the photophysics of polymethine fluorescent dyes in biomolecular environments. Q. Rev. Biophys. 44, 123–151 (2011).

    Article  CAS  PubMed  Google Scholar 

  22. Sauer, M. & Heilemann, M. Single-molecule localization microscopy in eukaryotes. Chem. Rev. 117, 7478–7509 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Jradi, F. M. & Lavis, L. D. Chemistry of photosensitive fluorophores for single-molecule localization microscopy. ACS Chem. Biol. 14, 1077–1090 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Gorka, A. P., Nani, R. R. & Schnermann, M. J. Cyanine polyene reactivity: scope and biomedical applications. Org. Biomol. Chem. 13, 7584–7598 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hernot, S., van Manen, L., Debie, P., Mieog, J. S. D. & Vahrmeijer, A. L. Latest developments in molecular tracers for fluorescence image-guided cancer surgery. Lancet Oncol. 20, e354–e367 (2019).

    Article  CAS  PubMed  Google Scholar 

  26. Barth, C. W. & Gibbs, S. L. Fluorescence image-guided surgery–a perspective on contrast agent development. Proc. SPIE Int. Soc. Opt. Eng. https://doi.org/10.1117/12.2545292 (2020).

  27. Feng, Z. et al. Excretable IR-820 for in vivo NIR-II fluorescence cerebrovascular imaging and photothermal therapy of subcutaneous tumor. Theranostics 9, 5706–5719 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wu, D. et al. Extrahepatic cholangiography in near-infrared II window with the clinically approved fluorescence agent indocyanine green: a promising imaging technology for intraoperative diagnosis. Theranostics 10, 3636–3651 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhu, S. et al. Near-infrared-II (NIR-II) bioimaging via off-peak NIR-I fluorescence emission. Theranostics 8, 4141–4151 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hu, Z. et al. First-in-human liver-tumour surgery guided by multispectral fluorescence imaging in the visible and near-infrared-I/II windows. Nat. Biomed. Eng. 4, 259–271 (2020).

    Article  PubMed  Google Scholar 

  31. Wang, S. et al. Anti-quenching NIR-II molecular fluorophores for in vivo high-contrast imaging and pH sensing. Nat. Commun. 10, 1058 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Cosco, E. D. et al. Flavylium polymethine fluorophores for near- and shortwave infrared imaging. Angew. Chem. Int. Ed. Engl. 56, 13126–13129 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Cosco, E. D. et al. Shortwave infrared polymethine fluorophores matched to excitation lasers enable noninvasive, multicolour in vivo imaging in real time. Nat. Chem. https://doi.org/10.1038/s41557-020-00554-5 (2020).

  34. Li, B., Lu, L., Zhao, M., Lei, Z. & Zhang, F. An efficient 1,064-nm NIR-II excitation fluorescent molecular dye for deep-tissue high-resolution dynamic bioimaging. Angew. Chem. Int. Ed. Engl. 57, 7483–7487 (2018).

  35. Bricks, J. L., Kachkovskii, A. D., Slominskii, Y. L., Gerasov, A. O. & Popov, S. V. Molecular design of near-infrared polymethine dyes: a review. Dyes Pigm. 121, 238–255 (2015).

    Article  CAS  Google Scholar 

  36. Gorka, A. P., Nani, R. R. & Schnermann, M. J. Harnessing cyanine reactivity for optical imaging and drug delivery. Acc. Chem. Res. 51, 3226–3235 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Friedman, H. C. et al. Establishing design principles for emissive organic SWIR chromophores from energy gap laws. Chem. https://doi.org/10.1016/j.chempr.2021.09.001 (2021).

  38. Cosco, E. D., Lim, I. & Sletten, E. M. Photophysical properties of indocyanine green in the shortwave infrared region. ChemPhotoChem https://doi.org/10.1002/cptc.202100045 (2021).

  39. Nani, R. R., Kelley, J. A., Ivanic, J. & Schnermann, M. J. Reactive species involved in the regioselective photooxidation of heptamethine cyanines. Chem. Sci. 6, 6556–6563 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Stackova, L. et al. Deciphering the structure-property relations in substituted heptamethine cyanines. J. Org. Chem. 85, 9776–9790 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Luciano, M. P. et al. A nonaggregating heptamethine cyanine for building brighter labeled biomolecules. ACS Chem. Biol. 14, 934–940 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pauli, J. et al. Suitable labels for molecular imaging—influence of dye structure and hydrophilicity on the spectroscopic properties of IgG conjugates. Bioconjug. Chem. 22, 1298–1308 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Nishio, N. et al. Optimal dosing strategy for fluorescence-guided surgery with panitumumab-IRDye800CW in head and neck cancer. Mol. Imaging Biol. 22, 156–164 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wilson, R. H., Nadeau, K. P., Jaworski, F. B., Tromberg, B. J. & Durkin, A. J. Review of short-wave infrared spectroscopy and imaging methods for biological tissue characterization. J. Biomed. Opt. 20, 030901 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Cao, J. et al. Recent progress in NIR-II contrast agent for biological imaging. Front. Bioeng. Biotechnol. 7, 487 (2019).

    Article  PubMed  Google Scholar 

  46. Dreher, M. R. et al. Tumor vascular permeability, accumulation and penetration of macromolecular drug carriers. J. Natl Cancer Inst. 98, 335–344 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Onda, N., Kimura, M., Yoshida, T. & Shibutani, M. Preferential tumor cellular uptake and retention of indocyanine green for in vivo tumor imaging. Int. J. Cancer 139, 673–682 (2016).

    Article  CAS  PubMed  Google Scholar 

  48. Newton, A. D. et al. Optimization of second window indocyanine green for intraoperative near-infrared imaging of thoracic malignancy. J. Am. Coll. Surg. 228, 188–197 (2019).

    Article  PubMed  Google Scholar 

  49. Ishizawa, T., Saiura, A. & Kokudo, N. Clinical application of indocyanine green-fluorescence imaging during hepatectomy. Hepatobiliary Surg. Nutr. 5, 322–328 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Harris, J., Kajdacsy-Balla, A. & Chiu, B. Creation of a murine orthotopic hepatoma model with intra-abdominal metastasis. Gastroenterol. Hepatol. Bed Bench 9, 174–179 (2016).

    PubMed  PubMed Central  Google Scholar 

  51. Troyan, S. L. et al. The FLARE intraoperative near-infrared fluorescence imaging system: a first-in-human clinical trial in breast cancer sentinel lymph node mapping. Ann. Surg. Oncol. 16, 2943–2952 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Tian, R. et al. Multiplexed NIR-II probes for lymph node-invaded cancer detection and imaging-guided surgery. Adv. Mater. 32, e1907365 (2020).

    Article  PubMed  Google Scholar 

  53. Usama, S. M., Inagaki, F., Kobayashi, H. & Schnermann, M. J. Norcyanine-carbamates are versatile near-infrared fluorogenic probes. J. Am. Chem. Soc. 143, 5674–5679 (2021).

    Article  CAS  PubMed  Google Scholar 

  54. Hatami, S. et al. Absolute photoluminescence quantum yields of IR26 and IR-emissive Cd1−xHgxTe and PbS quantum dots—method- and material-inherent challenges. Nanoscale 7, 133–143 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Würth, C., Grabolle, M., Pauli, J., Spieles, M. & Resch-Genger, U. Relative and absolute determination of fluorescence quantum yields of transparent samples. Nat. Protoc. 8, 1535–1550 (2013).

    Article  PubMed  Google Scholar 

  56. Nani, R. R. et al. In vivo activation of duocarmycin-antibody conjugates by near-infrared light. ACS Cent. Sci. 3, 329–337 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Institute for Laboratory Animal Research. Guide for the Care and Use of Laboratory Animals. 8th edn (National Academies Press, Washington, DC, 2011).

Download references

Acknowledgements

This work was supported by the Intramural Research Program of the National Institutes of Health including through a NCI FLEX award to M.J.S. (BC011506). We also acknowledge funding from the Helmholtz Zentrum München, the DFG—Emmy Noether program (no. BR 5355/2-1) and from the CZI Deep Tissue Imaging (DTI-0000000248) to O.T.B. U.R.-G. gratefully acknowledges funding from the DFG grant SIREN (DFG RE 1203/38-1). This project has also been funded in part with Federal funds from the National Cancer Institute, National Institutes of Health, under contract no. HHSN261200800001E. M.J.S. thanks J. Barchi, NCI-CCR, for nuclear magnetic resonance assistance and J. Kelley, NCI-CCR, for mass spectrometric analysis. M.S. and O.T.B. thank A. Feuchtinger, Helmholtz Munich imaging and pathology core facility, for support with the H&E staining of the cryosections. We also thank S. Glasl, P. Anzenhofer, U. Klemm, M. Warmer, S. Ramakrishnan for the excellent technical support. We also thank T. Klein, H. Robielski and B. Arus for helpful discussion.

Author information

Authors and Affiliations

Authors

Contributions

V.G.B., M.J.S. and O.T.B. designed the project. M.J.S. and O.T.B. jointly advised the project. M.J.S. and J.I. designed and carried out the computational chemistry. V.G.B. and M.P.L. performed organic synthesis and characterization. B.R. and U.R.-G. performed photophysical measurements and analyzed the data. T.S.B., J.G.P.L., M.S. and M.P.L. built the optical imaging setup. M.P.L. and N.L.P. performed phantom imaging experiments. M.N.N. and M.S. performed cell culture. M.S., C.S., C.M.R., L.R. and S.D. established animal tumor models and were responsible for animal handling and care. V.G.B., M.P.L. and M.S. performed bioconjugation chemistry and characterization. M.P.L., M.S., J.D.K., N.L.P. and J.G.P.L. designed and carried out animal imaging experiments. P.T.T. developed the imaging software. M.P.L. and M.S. processed and analyzed the imaging data. M.P.L. and M.J.S. wrote the manuscript. All authors have reviewed and approved the final version of the manuscript. Requests for materials and other correspondence should be addressed to M.J.S. and O.T.B.

Corresponding authors

Correspondence to Oliver T. Bruns or Martin J. Schnermann.

Ethics declarations

Competing interests

V.G.B., M.J.S., J.G.P.L., M.S. and O.T.B. have filed patent applications whose value may be affected by this publication. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks Samuel Achilefu and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Rita Strack was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Photostability.

Photostability of FNIR-866 and FNIR-1072 compared with IR-800CW (2 μM in 50 mM PBS, pH 7.4) using 20 mW/cm2 irradiation at 780 nm, 850 nm and 1064 nm, respectively. Experiments were run in quadruplicate and plotted as mean values with error bars derived from the standard deviation.

Source data

Extended Data Fig. 2 Chemical stability.

Chemical stability of IR-800CW, FNIR-866, FNIR-1072 (2 μM in 10% FBS/PBS) at room temperature. Experiments were run in quadruplicate and plotted as mean values with error bars derived from the standard deviation.

Source data

Extended Data Fig. 3 Phantom imaging.

Tail emission phantom comparison of 5 μM solutions of heptamethine (green bars) and nonamethine cyanines (cyan and purple bars) under various solvent and filter conditions when excited with their respective laser lines (785 nm, 890 nm, 1064 nm) at a power density of 90 mW/cm2.

Source data

Extended Data Fig. 4 Ex-vivo histological validation of tumor labeling after i.v. injection of IRDye800CW-Pan.

An athymic nude mouse bearing a MDA-MB-468 tumor (n = 1) was injected with 100 ug of IR-800CW-Pan (DOL 1.9, 1.2 nmol IR-800CW). Mouse was sacrificed 48 h after injection and the organs excised and imaged ex vivo (See Supplementary Figure 7). The tumor tissue was snap frozen in liquid nitrogen and 15 mm sections were cut. Adjacent sections were either stained with Hematoxylin/Eosin (H&E) for imaging in transmission or mounted with a DAPI containing mounting medium (ProLong™ Diamond Antifade Mountant with DAPI - ThermoFisher P36971) for imaging fluorescence. To detect fluorescence of IR-800CW and DAPI the section was imaged on a fluorescence microscope equipped with silicon based sCMOS camera and a customized excitation path coupling in a 785 nm laser for excitation. Overview images of the whole tumor section were captured by using the stitching routine of the microscope software using a 20X objective for IR-800CW-Pan and a 4X objective for DAPI. These are shown together with the H&E stained adjacent section in the top row. A magnified view including a central necrotic area and surrounding tumor cells was selected and images of this region are shown in the middle row for DAPI and IR-800CW-Pan. For visualizing the antibody signal at cellular resolution, images for DAPI and IR-800CW -Pan were also taken with a 20X objective and displayed individually and as colored overlay in the bottom row. Scale bars are displayed in the IR-800CW-Pan images. As expected the staining after in vivo labelling with the antibody is heterogeneous. The necrotic areas which can be seen in the H&E stained section are not labeled and the IR-800CW-Pan staining is more pronounced in the tumor rim and vital parts of the tumor. The images with 20X objective achieve cellular resolution and the staining of IR-800CW-Pan is localized on the cell surface. Images shown are representative from n = 3 slides.

Extended Data Fig. 5 Ex-vivo histological validation of tumor labeling after i.v. injection of FNIR-872-Pan.

An athymic nude mouse bearing a MDA-MB-468 tumor (n = 1) was injected with 114 ug of FNIR-872-Pan (DOL 1.1, 0.8 nmol FNIR-872). Mouse was sacrificed 48 h after injection and the organs excised and imaged ex vivo (See Supplementary Figure 7). The tumor tissue was snap frozen in liquid nitrogen and 15 mm sections were cut. Adjacent sections were either stained with Hematoxylin/Eosin (H&E) for imaging in transmission or mounted with a DAPI containing mounting medium (ProLong™ Diamond Antifade Mountant with DAPI - ThermoFisher P36971) for imaging fluorescence. The fluorescence of FNIR-872 and DAPI was imaged on a fluorescence microscope equipped with silicon based sCMOS camera and a customized excitation path coupling in a 785 nm laser for excitation. Overview images of the whole tumor section were captured by using the stitching routine of the microscope software using a 20X objective for FNIR-872-Pan and DAPI. These are shown together with the H&E stained adjacent section in the top row. A magnified image including a central necrotic area and surrounding tumor cells was selected and images of this region are shown in the middle row DAPI and FNIR-872-Pan. For visualizing antibody signal at cellular resolution, images for DAPI and FNIR-872-Pan were also taken with a 20X objective and displayed individually and as colored overlay in the bottom row. Scale bars are displayed in the FNIR-872-Pan images. As expected the staining after in vivo labelling with the antibody is heterogeneous. The necrotic areas which can be seen in the H&E stained section are not labeled and the FNIR-872-Pan staining is more pronounced in the tumor rim and vital parts of the tumor. The images with 20X objective achieve cellular resolution and the staining of FNIR-872-Pan is localized on the cell surface. Images shown are representative from n = 3 slides.

Extended Data Fig. 6 Control for ex-vivo histological validation of tumor labeling after i.v. injection of IRDye800CW-Pan and FNIR-872-Pan.

An athymic nude mouse bearing a MDA-MB-468 tumor (n = 1) was sacrificed without injection of a contrast agent and the organs excised. The tumor tissue was snap frozen in liquid nitrogen and 15 mm sections were cut. Adjacent sections were either stained with Hematoxylin/Eosin (H&E) for imaging in transmission or mounted with a DAPI containing mounting medium (ProLong™ Diamond Antifade Mountant with DAPI - ThermoFisher P36971) for imaging fluorescence. The same settings as IR-800CW and FNIR-872 were applied and DAPI was imaged on a fluorescence microscope equipped with silicon based sCMOS camera and a customized excitation path coupling in a 785 nm laser for excitation. Overview images of the whole tumor section were captured by using the stitching routine of the microscope software. These are shown together with the H&E stained adjacent section in the top row. A magnified view including a central necrotic area and surrounding tumor cells was selected and images of this region are shown in the middle row for DAPI and the 785 nm excitation channel. For visualization with cellular resolution, images for DAPI and the 785 nm excitation channel were also taken with a 20X objective and displayed individually and as colored overlay in the bottom row with the same display settings as for FNIR-872-Pan. Scale bars are displayed in the 785 nm excitation channel images. There is no significant signal in the 785 nm excitation channel.

Extended Data Fig. 7 2-Color Imaging with FNIR-Tag-Dextran and FNIR-872-Pan.

Two color imaging using FNIR-Tag-Dex (10 kDa, 5 nmol effective dye in 100 μL PBS) and FNIR-872-Pan (100 μg, DOL 1.4, 1 nmol effective dye in 100 μL PBS) in an athymic nude mouse bearing an MDA-MB-468 xenograft tumor implanted s.c. in the right flank (n = 1). Imaging settings: Images were acquired on a NIRvana 640ST using 3×1150 long pass filters. The lasers (785 nm for FNIR-Tag-Dex and 890 nm for FNIR-872-Dex) were power matched at 100 mW/cm2 measured over the tumor for each imaging timepoint. Camera exposure times were 75 ms and 400 ms for the FNIR-Tag-Dex and FNIR-872-Pan channels, respectively. For in vivo images, the color scale is from 0-273 and 14-50 counts/ms in the 785 and 890 channel, respectively. For the ex vivo images, the color scale is from 0-440 and 7-88 counts/ms in the 785 and 890 channel, respectively.

Extended Data Fig. 8 Single Color Imaging of Hep3B Orthotopic Liver Tumors.

Side by side comparison of NIR (a) (Perkin-Elmer IVIS) and SWIR (b) detection of a representative Hep3B orthotopic bearing or naïve athymic nude mice after receiving ICG (2 mg/kg). Images were recorded at 72 h postinjection. SWIR detection enables multiplexing of a marker for healthy liver tissue (FNIR-872-Dex) to aid in delineation of the tumor margins (see Fig. 3a). Plot profiles for Hep3B and naïve mice (c, IVIS and d, SWIR), drawn as indicated by the red lines, demonstrate the enhanced resolution of imaging in the SWIR.

Source data

Extended Data Fig. 9 Crosstalk between ICG and FNIR-872-Pan in vivo.

An athymic nude mouse bearing an MDA-MB-468 xenograft tumor implanted orthotopic in the mammary fat pad was imaged before injection (prescan) of FNIR872-Pan (140 μg, DOL 1.1, 1.1 nmol effective dye in 100 μL PBS, i.v.). The mouse was imaged again one day after injection of FNIR872-Pan (pre ICG injection) and ca 1 hour after injection of ICG intradermal (ca. 100 nmol ICG in water) at the base of the tail (post ICG injection). The signal of FNIR872-Pan was calculated as the difference of the intensity in the 890 nm channel (pre ICG injection – prescan) at the tumor. Crosstalk of FNIR872-Pan in the 785 nm channel was calculated as the difference of the intensity in the 785 nm channel (pre ICG injection – prescan) at the tumor. The signal of ICG was calculated as the difference of the intensity in the 785 nm channel (post ICG injection – pre ICG injection) at the lymphatic vessels closed to the tumor. Crosstalk of ICG in the 890 nm channel was calculated as the difference of the intensity in the 890 nm channel (post ICG injection – pre ICG injection) at the same lymphatic vessels. Crosstalk between ICG and FNIR-872-Pan is very small with a signal to crosstalk ratio of around 12 for the 785 nm ICG channel and a signal to crosstalk ratio of around 17 for the 890 nm FNIR-872-Pan channel. Imaging settings: Images were acquired with Allied Vision G-32 camera using 2X 1000 nm long pass filters and 3X 1050 nm long pass filters. Laser 785 nm (prescan dark-subtracted image taken with 50 ms exposure time and divided by a factor of 5 and dark-corrected pre-ICG injection image taken with 25 ms exposure time and divided by a factor of 2.5 to match the time-point after injection of ICG taken with 10 ms exposure time, 80 mW/cm2) and laser 890 nm (prescan dark-subtracted images were taken with 50 ms exposure time and divided by a factor of 2 to match the time-points after injection of Pan-FNIR872 taken with 25 ms exposure time, 115 mW/cm2) were sequentially triggered by CCDA software together with the 1300 nm LED (10 ms exposure time) as reflection channel. MFI of 785 nm channel is displayed in counts/10 ms, whereas MFI of 890 nm channel is displayed in counts/25 ms.

Extended Data Fig. 10 Crosstalk between ICG and FNIR-1072-Pan in vivo.

An athymic nude mouse bearing an MDA-MB-468 xenograft tumor implanted orthotopic in the mammary fat pad was imaged before injection (prescan) of FNIR-1072-Pan (103 μg, DOL 0.53, 0.4 nmol effective dye in 100 μL PBS, i.v.). The mouse was imaged again one day after injection of FNIR1072-Pan (pre ICG injection) and 2 hour after injection of ICG intradermal (ca. 100 nmol ICG in water) at the base of the tail (post ICG injection). The signal of FNIR1072-Pan was calculated as the difference of the intensity in the 968 nm channel (pre ICG injection – prescan) at the tumor. Crosstalk of FNIR1072-Pan in the 785 nm channel was calculated as the difference of the intensity in the 785 nm channel (pre ICG injection – prescan) at the tumor. The signal of ICG was calculated as the difference of the intensity in the 785 nm channel (post ICG injection – pre ICG injection) at the lymph node. Crosstalk of ICG in the 968 nm channel was calculated as the difference of the intensity in the 968 nm channel (post ICG injection – pre ICG injection) at the lymph node. Crosstalk between ICG and FNIR-1072-Pan is very small with a signal to crosstalk ratio of around 15 for the 785 nm ICG channel and a signal to crosstalk ratio of around 140 for the 968 nm FNIR-1072-Pan channel. Imaging settings: images were acquired with Allied Vision G-32 camera using 2X 1000 nm long pass filters and 3X 1050 nm long pass filters. Laser 785 nm (prescan dark corrected images taken with 100 ms exposure time and divided by a factor of 4 to match the time-points after injection of Pan-FNIR1072 taken with 25 ms exposiure time, 80 mW/cm2) and 968 nm (dark subtracted images were taken with 100ms exposure time and divided by a factor of 2 to match the time-points after injection, 350 mW/cm2) were sequentially triggered by CCDA software together with the 1300 nm LED (20 ms exposure time) as reflection channel. MFI of 785 nm channel is displayed in counts/25 ms whereas MFI of 968 nm channel is displayed in counts/50 ms.

Supplementary information

Supplementary Information

Supplementary Figs. 1–9 and Supplementary Notes 1–4

Reporting Summary

Supplementary Video 1

Imaging of an awake mouse 24 h after injection of panitumumab-FNIR-872 for tumor targeting. Reflected light images of the mouse were obtained using an LED at 1,300 nm. Image sequence was acquired with a frame rate of 7 frames per second (f.p.s.).

Supplementary Video 2

Real-time multiplexed in vivo visualization in three colors of lymphatics pumping and lymph nodes in a tumor-bearing mouse 24 h after injection of panitumumab-FNIR-872 for tumor labeling and 30 min after injection of ICG intradermally (i.d.) before injection of dextran-FNIR-1072. Reflected light images of the mouse were obtained using an LED at 1,300 nm. Image sequence was acquired with a frame rate of 4 f.p.s.

Supplementary Video 3

Real-time multiplexed in vivo imaging in four colors of the injection of dextran-FNIR-1072 in a tumor-bearing mouse 24 h after injection of panitumumab-FNIR-872 for tumor labeling and 1 h after injection of ICG (i.d.) for lymphatics and lymph node visualization. Reflected light images of the mouse were obtained using an LED at 1,300 nm. Image sequence was acquired with a frame rate of 4 f.p.s.

Supplementary Video 4

Real-time multiplexed fluorescence-guided dissection of tumor in a mouse injected with panitumumab-FNIR-872 (i.v.), ICG (i.d.) and dextran-FNIR-1072 (i.v.). Reflected light images of the mouse were obtained using an LED at 1,300 nm. Image sequence was acquired with a frame rate of 4 f.p.s.

Supplementary Video 5

Real-time multiplexed fluorescence-guided dissection of lymph nodes in a mouse injected with panitumumab-FNIR-872 (i.v.), ICG (i.d.) and dextran-FNIR-1072 (i.v.). Reflected light images of the mouse were obtained using an LED at 1,300 nm. Image sequence was acquired with a frame rate of 4 f.p.s.

Supplementary Video 6

Real-time multiplexed fluorescence-guided dissection of tumor in a mouse injected with panitumumab-FNIR-1072 (i.v.), ICG (i.d.) and dextran-FNIR-872 (i.v.). Reflected light images of the mouse were obtained using an LED at 1,300 nm. Image sequence was acquired with a frame rate of 6 f.p.s.

Supplementary Video 7

Real-time multiplexed fluorescence-guided dissection of lymph nodes in a mouse injected with panitumumab-FNIR-1072 (i.v.), ICG (i.d.) and dextran-FNIR-872 (i.v.). Reflected light images of the mouse were obtained using an LED at 1,300 nm. Image sequence was acquired with a frame rate of 6 f.p.s.

Source data

Source Data Fig. 1

Singlet oxygen generation quantum yield kinetics.

Source Data Fig. 2

Tumor-to-background data.

Source Data Fig. 3

Plot profile data.

Source Data Extended Data Fig. 1

Photostability kinetic data.

Source Data Extended Data Fig. 2

Chemical stability kinetic data.

Source Data Extended Data Fig. 3

Tail emission quantification.

Source Data Extended Data Fig. 8

Hep3B plot profile data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bandi, V.G., Luciano, M.P., Saccomano, M. et al. Targeted multicolor in vivo imaging over 1,000 nm enabled by nonamethine cyanines. Nat Methods 19, 353–358 (2022). https://doi.org/10.1038/s41592-022-01394-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-022-01394-6

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer