Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2023 Sep 4:18:4949-4967.
doi: 10.2147/IJN.S425013. eCollection 2023.

Multifunctional Near-Infrared Dye IR-817 Encapsulated in Albumin Nanoparticles for Enhanced Imaging and Photothermal Therapy in Melanoma

Affiliations

Multifunctional Near-Infrared Dye IR-817 Encapsulated in Albumin Nanoparticles for Enhanced Imaging and Photothermal Therapy in Melanoma

Jianv Wang et al. Int J Nanomedicine. .

Abstract

Background: Near-infrared cyanine dyes have high sensitivity and spatial resolution imaging capabilities, but they also have unavoidable drawbacks such as photobleaching, low water solubility, fluorescence quenching, and toxic side effects. As an effective biologic drug carrier, albumin combines with cyanine dyes to form albumin@dye nanoparticles. These nanoparticles can alleviate the aforementioned issues and are widely used in tumor imaging and photothermal therapy.

Methods: Herein, a newly synthesized near-infrared dye IR-817 was combined with bovine serum albumin (BSA) to create BSA@IR-817 nanoparticles. Through the detection of fluorescence emission and absorption, the optimal concentration and ratio of BSA and IR-817 were determined. Subsequently, dynamic light scattering (DLS) measurements and scanning electron microscopy (SEM) were used for the physical characterization of the BSA@IR-817 nanoparticles. Finally, in vitro and in vivo experiments were conducted to assess the fluorescence imaging and photothermal therapeutic potential of BSA@IR-817 nanoparticles.

Results: IR-817 was adsorbed onto the BSA carrier by covalent conjugation and supramolecular encapsulation, resulting in the formation of dispersed, homogeneous, and stable nanoparticles with a particle size range of 120-220 nm. BSA@IR-817 not only improved the poor water solubility, fluorescence quenching, and toxic side effects of IR-817 but also enhanced the absorption and fluorescence emission peaks in the near-infrared region, as well as the fluorescence in the visible spectrum. In addition, BSA@IR-817 combined with laser 808 irradiation was able to convert light energy into heat energy with temperatures exceeding 50 °C. By creating a mouse model of subcutaneous melanoma, it was discovered that the tumor inhibition rate of BSA@IR-817 was greater than 99% after laser irradiation and that it achieved nearly complete tumor ablation without causing significant toxicity.

Conclusion: Our research, therefore, proposes the use of safe and effective photothermal nanoparticles for the imaging, diagnosis, and treatment of melanoma, and offers a promising strategy for future biomedical applications.

Keywords: albumin; cyanine dyes; melanoma; near-infrared imaging; photothermal therapy.

PubMed Disclaimer

Conflict of interest statement

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Figures

Scheme 1
Scheme 1
Schematic illustration of BSA@IR-817 for NIR fluorescence image and photothermal therapy of melanoma. By Figdraw.
Figure 1
Figure 1
Rational screening process of HSA@IR-817 and BSA@IR-817. Fluorescence emission spectra of different concentrations of (A) BSA@IR-817 and (B) HSA@IR-817 with the molar ratio of albumin/IR-817 as 1:1 in PBS. Fluorescence emission spectra of 10 μM IR-817 mixed with different concentrations of (C) BSA or (D) HSA to achieve different ratios (4:1, 3:1, 2:1, 1:1, 1:2, 1:3, and 1:4). Electrophoresis gel analysis of (E) BSA@IR-817 and (F) HSA@IR-817 in NIR and white light. (G) Infrared absorption spectra of BSA@IR-817 and HSA@IR-817 (10 μM, 1:1). (H) NIR image of 10 μM BSA@IR-817 and IR-817 in an EP tube. (I) Infrared absorption spectra and (J) fluorescence emission spectra of 10 μM BSA@IR-817, BSA@ICG, IR-817 in BSA, IR-817 in PBS, ICG in BSA, and ICG in PBS.
Figure 2
Figure 2
Characterization of BSA@IR-817 nanoparticles. (A) Scanning electron microscopy (SEM) image of BSA@IR-817. Scale bars: 1 μm (left image); 200 nm (right image). (B) Hydrodynamic size distribution of BSA and BSA@IR-817. (C) The hydrodynamic diameter of BSA@IR-817 during 12 days. (D) Zeta potential of BSA@IR-817, BSA, and IR-817. (E) The relative fluorescence intensity of BSA@IR-817 and IR-817 at different concentrations. (F) The relative fluorescence intensity of 10 μM BSA@IR-817, IR-817, and ICG right after the dilution process or 24 h of storage (in light or dark).
Figure 3
Figure 3
The fluorescence imaging of BSA@IR-817 in vitro and in vivo. (A) The fluorescence images of B16-F10 cells were incubated with 10 μM BSA@IR-817, IR-817, and ICG for different times (0.5, 1, 2, 4, 8, and 16 h). (B) The quantification of relative fluorescence signal intensity in (A) changed over time. (C) Fluorescence imaging of the mice at different time points (0, 2, 6, 12, and 24 h) after intravenous injection of BSA@IR-817, IR-817, and ICG. The white dashed circle indicated the inoculated tumor area in the mice. (D) Fluorescence statistics of BSA@IR-817, IR-817, and ICG in mice. Values represent the mean ± SD, * p <0.05, *** p <0.001, **** p <0.0001, compared with the other two groups.
Figure 4
Figure 4
Evaluations BSA@IR-817 photothermal properties. (A) Temperature increase of BSA@IR-817 (20 μM) in PBS under an 808 nm laser with different exposure intensities (0.5, 0.8, 1 W/cm2). (B) Temperature increase of BSA@IR-817 at different concentrations (0, 5, 10, and 20 μM) under an 808 nm laser (1 W/cm2). (C) Temperature change of BSA@IR-817, IR-817, ICG and BSA (20 μM) upon exposure to 808 nm laser (1 W/cm2) for different intervals. (D) Infrared thermal images of (C). (E) Temperature changes of BSA@IR-817, IR-817 and ICG during four circles of heating-cooling processes (1 W/cm2, 5 min). (F) Photothermal conversion efficiency (PCE) of BSA@IR-817, IR-817 and ICG (20 μM, 1 W/cm2). (G) Infrared thermographic images of the B16-F10 cell tumor-bearing mice after irradiation (1 W/cm2, 5 min). (H) Change in tumor temperature as a function of the irradiation time.
Figure 5
Figure 5
Photoinduced cytotoxicity of BSA@IR-817 in melanoma cells. (A) Viabilities of cancer cells incubated with BSA@IR-817 for 24 h. (B) Photoinduced toxicity of cancer cells treated with BSA@IR-817 of different concentrations following 808 nm laser irradiation (1 W/cm2) for 5 min and continued incubation for 24 h. (C) Fluorescence images of Calcein AM/PI co-stained A375 and B16-F10 cells with BSA@IR-817 (10 μM) incubation after being exposed to the 808 nm laser (1 W/cm2, 5 min). Values represent the mean ± SD, * p <0.05, ** p <0.01, *** p <0.001, **** p <0.0001, compared with the control group.
Figure 6
Figure 6
Photo-therapeutic efficacy evaluation of BSA@IR-817 in vivo. (A) Establishment of B16-F10 tumor-bearing mice model, administration, and laser irradiation to the mice. (B) Photos of mice bearing B16-F10 tumor and anatomic tumor on day 10 after various treatments as indicated. (C) The tumor growth in B16-F10 tumor-bearing mice treated by different formulations. (D) Tumor weight. (E) The tumor growth curves of each group. (F) Body weight changes of mice. (G) Blood safety evaluation of different concentrations of BSA@IR-817. (H) The survival curves of the tumor-bearing mice. Values represent the mean ± SD, *p <0.05, **p <0.01, ***p <0.001, ****p <0.0001.
Figure 7
Figure 7
Histological analysis. (A) Representative histological features of the major organs of the tumor-bearing mice with various treatments. Scale bars, 50 μm (B) Ki-67, CC-3, Bax, and Bcl-2 immunohistochemical staining of tumor sections in the mice. Scale bars, 50 μm. Values represent the mean ± SD, n = 3. **p <0.01, ***p <0.001, ****p <0.0001.

Similar articles

Cited by

References

    1. Guo W, Wang H, Li C. Signal pathways of melanoma and targeted therapy. Signal Transduct Target Ther. 2021;6(1):424. doi:10.1038/s41392-021-00827-6 - DOI - PMC - PubMed
    1. Hsu CH, Lee KJ, Chiu YH, et al. The lysosome in malignant melanoma: biology, function and therapeutic applications. Cells. 2022;11(9):1492. doi:10.3390/cells11091492 - DOI - PMC - PubMed
    1. Wagstaff W, Mwamba RN, Grullon K, et al. Melanoma: molecular genetics, metastasis, targeted therapies, immunotherapies, and therapeutic resistance. Genes Dis. 2022;9(6):1608–1623. doi:10.1016/j.gendis.2022.04.004 - DOI - PMC - PubMed
    1. Long GV, Menzies AM, Scolyer RA. Neoadjuvant checkpoint immunotherapy and melanoma: the time is now. J Clin Oncol. 2023;O2202575. - PubMed
    1. Serrati S, Guida M, Di Fonte R, et al. Circulating extracellular vesicles expressing PD1 and PD-L1 predict response and mediate resistance to checkpoint inhibitors immunotherapy in metastatic melanoma. Mol Cancer. 2022;21(1):20. doi:10.1186/s12943-021-01490-9 - DOI - PMC - PubMed

Grants and funding

This work was supported by the National Natural Science Foundation of China (grant number: 82003716), Sichuan Science and Technology Program (2022YFS0631, 2022YFS0635), Luzhou Science and Technology Projects (2021-JYJ-48, 2022-JYJ-141), Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (Number: ZYYCXTD-C-202207), Southwest Medical University School-level Project Fund (2021ZKMS049, 2022ZD011).
-