You are on page 1of 6

Journal of Photochemistry & Photobiology, B: Biology 241 (2023) 112674

Contents lists available at ScienceDirect

Journal of Photochemistry & Photobiology, B: Biology


journal homepage: www.elsevier.com/locate/jphotobiol

Unadulterated BODIPY nanoparticles as light driven antibacterial agents for


treating bacterial infections and promoting wound healing
Qiaoxia Shi a, Xinyuan Wang a, Hongxin Liu a, Zhigang Xie b, Min Zheng a, *
a
School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun, Jilin 130012, PR
China
b
State Key Laboratory of Polymer Chemistry and Physics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, Jilin
130022, PR China

A R T I C L E I N F O A B S T R A C T

Keywords: Antimicrobial photodynamic therapy (aPDT) is an effective strategy to eliminate bacteria without inducing
Antimicrobial photodynamic therapy bacterial resistance. As typical aPDT photosensitizers, most of boron-dipyrromethene (BODIPY) are hydrophobic,
Boron-dipyrromethene and nanometerization is imperative to render them dispersible in physiological media. Recently, carrier-free
Carrier-free
nanoparticles (NPs) are formed via the self-assembly of BODIPYs without the help of any surfactants or auxil­
Self-assembly
Nanoparticle
iaries, arousing people’s interest. So as to fabricate carrier-free NPs, BODIPYs usually need to be derived into
Wound healing dimers, trimers, or amphiphiles through complex reactions. Few unadulterated NPs were obtained from BODIPYs
with precise structures. Herein, BNP1-BNP3 were synthesized by the self-assembly of BODIPY, which showed
excellent anti-Staphylococcus aureus ability. Among them, BNP2 could effectively fight bacterial infections and
promote wound healing in vivo.

1. Introduction solvents and then diluted with aqueous medium. However, the strong
hydrophobicity leads to the precipitation from water solution often oc­
Bacterial infections are a serious threat to human health [1–4], curs, which is the biggest obstacles for their biomedical applications
hitherto, although lots of antibiotics have been developed for the [26,27]. One feasible way to solve this problem is the nanometerization
treatment of bacteria-caused diseases. The overuse of antibiotics greatly of small molecules by the assembly with polymers [28–30]. However,
increases the bacterial resistance, which brings great difficulties to cure the shortcomings including inevitable toxicity of carriers, uncontrolled
numerous infectious diseases [5]. To address this issue, it is still a great leakage of dyes and poor experimental reproducibility are hard to
challenge to explore antibiotic-free therapeutic mode to inactivate overcome [31–33].
pathogens [6,7]. Antimicrobial photodynamic therapy (aPDT) is an Recently, the carrier-free strategy that constructing organic nano­
effective method that can eradicate bacterial by utilizing photosensi­ particles (NPs) via the self-assembly of small molecules has attracted
tizers (PSs) to generate toxic reactive oxygen species (ROS) under the increasing attention, which possesses the advantages of almost 100%
illumination of suitable light [8]. loading rate, precise molecular structures, good colloidal stability and
Boron-dipyrromethene (BODIPY) and its derivatives are a kind of repeatable synthesis [34]. In order to construct carrier-free NPs, BODI­
robust dyes, which have attracted extensive attention because of their PYs have to be derived into dimers [35–38], trimers [39], or amphi­
abundant chemical structures, good photostability, easy modification philes [40–42] via complicated synthesis steps. As far as we know, rare
and excellent optical properties [9,10]. Many BODIPY dyes have been BODIPYs with simple structures can form NPs without any surfactants or
successfully applied in sensing [11–13], bioimaging [14–17], and cancer auxiliaries [43,44].
treatment [18–21]. Especially, some BODIPYs are promising PSs for Herein, a BODIPY derivative (Scheme 1) could self-assemble into
photodynamic therapy of tumor [22]. By contrast, just a little work on NPs (BNP1-BNP3) via a convenient nanoprecipitation method [45–47].
photodynamic inactivation of bacteria have been reported [23–25]. Among them, BNP2 had good water solubility, stability and cyto­
Due to the inherent hydrophobicity, most of the BODIPY dyes are compatibility, which could eradicate Staphylococcus aureus (S. aureus) by
insoluble in physiological medium, they need to be dissolved in organic producing the singlet oxygen (1O2) under green LED light. The potent

* Corresponding author.
E-mail address: zhengm@ciac.ac.cn (M. Zheng).

https://doi.org/10.1016/j.jphotobiol.2023.112674
Received 12 September 2022; Received in revised form 1 February 2023; Accepted 21 February 2023
Available online 24 February 2023
1011-1344/© 2023 Elsevier B.V. All rights reserved.
Q. Shi et al. Journal of Photochemistry & Photobiology, B: Biology 241 (2023) 112674

1
antibacterial activity enabled BNP2 to cure bacterial infections and O2 within the bacteria under green light irradiation.
accelerate wound healing.
2.3. Antibacterial Photodynamic Activity in Vitro
2. Results and Discussion
On the basis of the above results, we investigated the antibacterial
2.1. Synthesis and Characterization photodynamic effects of BNP1-BNP3 (10 ng/mL) against S. aureus in
vitro. Without light irradiation, a large number of colonies grew on the
BODIPY was synthesized according to previous reports [9,48,49], plates of the BNP1-BNP3 treatment groups (Fig. 3A), the bacterial sur­
and BNP1-BNP3 were prepared by nanoprecipitation method. Take vival rates were about 98% (Fig. 3B), and the growth curves were similar
BNP2 as an example: the tetrahydrofuran (THF) solution of BODIPY was to the control group (Fig. 3C), proving that BNP1-BNP3 had no dark
added dropwise to water, stirred overnight until THF was completely toxicity to bacteria. As exposure to green LED light (18 mW cm− 2, 5
volatilized, then dialyzed against deionized water. The preparation min), the bacteriostatic effect of BNP1-BNP3 was very significant, no
procedures of BNP1 and BNP3 were similar to BNP2, except that the colonies grew on the plates, and there was still no amplification after 33
solvent used to dissolve BODIPY was acetone and acetone containing h (Fig. 3D), indicating they had excellent antibacterial activity.
1.5% (mass ratio) 1,2-propanediol, respectively. The concentration of Next, we assessed the photodynamic effects of BNP1-BNP3 on
BNPs was calculated according to the standard curve of BODIPY S. aureus biofilms by using crystal violet (CV) staining. As shown in
(Fig. S1). The hydrodynamic diameter (HD) of BNP1-BNP3 determined Fig. 4A, under the condition of no light, the BNP1-BNP3 treated groups
by dynamic light scattering (DLS) was 248.6, 224, and 188.3 nm, formed biofilms, so the biofilms on the wells of the 96-well plates were
respectively (Fig. 1A). The size of BNP2 remained almost unchanged stained purple by CV. In the presence of light (18 mW cm− 2, 5 min), the
after 120 d of storage (Fig. S2), indicating that BNP2 had good colloidal bacteria treated with 10 ng/mL of BNP1-BNP3 could not form biofilms,
stability. Transmission electron microscopy (TEM) image demonstrated and after the addition of CV, the wells of the 96-well plate remained
that BNP2 was quasi-spherical nanoparticle with an average size of colorless due to absence of biofilms. The biomass was as low as 1%
~120 nm (Fig. 1B), which was smaller than HD due to the shrinkage of (Fig. 4B), indicating that BNP1-BNP3 could effectively inhibit the for­
BNP2 in the solid state. The zeta potentials of BNP1-BNP3 were 0.288, mation of S. aureus biofilm.
0.207 and 0.021 mV, respectively. In the UV–vis absorption spectra of Then we selected BNP2 as the representative for Live/Dead staining
BNP1-BNP3 (Fig. 1C), an absorption maximum was observed at 543 nm, to study antibacterial mechanism. S. aureus were treated with PBS and
which was red-shifted by 8 nm relative to BODIPY (535 nm). BNP2, and stained with SYTO 9 (green fluorescence, live bacteria) and
propidium iodide (PI, red fluorescence, dead bacteria) after irradiating
2.2. Detection of 1O2 with green light, then were observed by CLSM. As shown in Fig. 4C, no
obvious red fluorescence was observed in PBS group, indicating that cell
1
O2 generation ability of BNPs aqueous solution was assessed by membrane was intact. While, S. aureus in BNP2 groups emitted intense
using indocyanine green (ICG). As shown in Fig. 1D and Fig. S3, in the red fluorescence, proving that cell membrane was damaged and the
presence of BNPs, the absorbance of ICG gradually decreased with the bacteria died. The above results further confirmed that BNP2 could
prolongation of green LED illumination time. After 160 s, the absor­ rapidly inactivate S. aureus by destroying cell membrane.
bance of ICG decreased by 41, 68 and 44% in BNP1-BNP3 groups,
respectively, implying that BNP2 had the highest 1O2 generation ability. 2.4. Antibacterial Activity of BNP2 In Vivo
BNP2 was selected as the typical representative to evaluate the 1O2-
generating capacity in S. aureus cells by using 1O2 detection kit (DCFH- The mouse wound infection model was established to evaluate the
DA). Bacteria were treated with PBS and BNP2, respectively. Bacteria in antibacterial activity of BNP2 in vivo. First, 12 female Kunming mice
the PBS + L and BNP2 + L groups were exposed to green LED light. were randomly divided into 4 groups. After the mouse was anesthetized,
Subsequent staining was performed according to the manufacturer’s circular wound with diameter about 0.8 cm was formed on the back
instructions and the results were observed using a confocal laser scan­ skin. Subsequently, wound infections of each mouse were induced with
ning microscope (CLSM). As depicted in Fig. 2, the BNP2 + L group S. aureus (1 × 107 CFU/mL), then were treated with PBS and BNP2 (350
showed bright green fluorescence, indicating that BNP2 could produce ng/mL). The light groups (PBS + L and BNP2 + L) were irradiated with

Scheme 1. Synthesis and antibacterial application of BNP2 in vivo.

2
Q. Shi et al. Journal of Photochemistry & Photobiology, B: Biology 241 (2023) 112674

Fig. 1. (A) HD of BNP1-BNP3. (B) TEM image of BNP2. (C) UV–vis absorption spectra of BNP1-BNP3 and BODIPY. (D) Time-dependent photobleaching of ICG
induced by 1O2 generated from BNP1-BNP3 (200 ng/mL) under green LED light (18 mW cm− 2). (For interpretation of the references to colour in this figure legend,
the reader is referred to the web version of this article.)

Fig. 2. CLSM images of intracellular 1O2 generation in S. aureus cells treated with PBS and BNP2 (10 ng/mL) in the absence or presence of green light (18 mW cm− 2,
5 min). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

green LED light (18 mW cm− 2) for 5 min. Wound photos were taken 2.5. Biosafety Assessment of BNP2
(Fig. 5A), the wound areas were measured to calculate the wound
healing rates (Fig. 5B), and mock images of wounds were drawn The biosafety of BNP2 was assessed by 4,5-dimethylthiazol-2-yl)-2,5-
(Fig. 5C). After 14 d of treatment, the wounds of mice in the BNP2 + L diphenyltetrazolium bromide (MTT) and hemolysis assay. First, mouse
group were almost completely healed. Their healing rates were as high fibroblasts (L929) were used to evaluate the cytocompatibility of BNP2
as 90.7%, and the bacteria on the wounds surface were negligible, which by MTT assay. As shown in Fig. 6C, when the concentration reached to
were sharp contrast with the other three treatment groups where there 1000 ng/mL, the survival rate of L929 was still greater than 90%,
were still large-sized wounds and a mass of bacteria on the wounds verifying BNP2 had good cytocompatibility. Next, hemolysis experiment
surface (Fig. 5D and E). During the whole treatment period, the weight was performed using mouse blood. Compared with the positive control
of mice in each group increased steadily (Fig. 6A), indicating that BNP2 (Triton X-100), the hemolytic capacity of BNP2 was negligible (Fig. 6D),
did not cause any adverse effect on mice and had good biological safety. confirming BNP2 had good hemocompatibility.
On the 14th day, the wound skin tissues of mice were stained with he­
matoxylin and eosin (H&E). As depicted in Fig. 6B, the wounds of mice 3. Conclusions
in the PBS, PBS + L and BNP2 groups had severe inflammation, while no
obvious inflammation was observed in the BNP2 + L group. In summary, carrier-free BNPs were synthesized via the self-
The wound tissues of BNP2 + L group regenerated blood vessels, and assembly of BODIPY, which had high ROS generation capacity. BNP1-
new hair follicles formed, indicating that the inflammation was signifi­ BNP3 exhibited excellent anti-S. aureus activity, which could kill bac­
cantly relieved. The results mentioned above demonstrated that the teria and inhibit the formation of biofilm. In addition, in vivo experi­
photodynamic antibacterial activity of BNP2 could effectively combat ments proved that BNP2 could cure bacterial infections of wounds and
bacterial infections of wounds in vivo and promote wound healing. accelerate wound healing. This work provides a new strategy of

3
Q. Shi et al. Journal of Photochemistry & Photobiology, B: Biology 241 (2023) 112674

Fig. 3. (A) Photographs and (B) survival rates of colony forming units (CFU) on solid plates after 10 ng/mL of BNP1-BNP3 treatment of S. aureus. Growth curves of
S. aureus treated with BNP1-BNP3 (10 ng/mL) without (C) or with (D) green light irradiation (18 mW cm− 2, 5 min). (For interpretation of the references to colour in
this figure legend, the reader is referred to the web version of this article.)

Fig. 4. (A) Photographs and (B) quantification of residual biofilms stained with CV. (C) CLSM images of Live/Dead staining of S. aureus after treating with PBS and
BNP2 under green light irradiation (18 mW cm− 2, 5 min). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version
of this article.)

constructing unadulterated NPs as effective PDT agents for biomedical Zhigang Xie: Resources, Supervision. Min Zheng: Conceptualization,
applications. Resources, Supervision, Writing – review & editing, Data curation.

CRediT authorship contribution statement Declaration of Competing Interest

Qiaoxia Shi: Methodology, Investigation, Software, Writing – orig­ The authors declare no conflicts of interest.
inal draft. Xinyuan Wang: Investigation. Hongxin Liu: Investigation.

4
Q. Shi et al. Journal of Photochemistry & Photobiology, B: Biology 241 (2023) 112674

Fig. 5. (A) Time-dependent images and (B) wound healing rates of wounds in different treatment groups, which were infected by S. aureus. (C) Mock images of
wounds within 14 d of treatment (1 d: red; 3 d: yellow; 5 d: green; 7 d: blue; 14 d: pink). (D) Photographs and (E) bacterial survival of colonies on solid plates from
wounds. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

Fig. 6. (A) Mice body weight changes during the treatment. (B) H&E staining images of wound skin tissues in different treatment groups (red arrows: blood vessels;
green arrows: hair follicles). (C) Cell viability of L929 treated with different concentrations of BNP2. (D) Hemolysis rate of BNP2 at 350 ng/mL. (For interpretation of
the references to colour in this figure legend, the reader is referred to the web version of this article.)

5
Q. Shi et al. Journal of Photochemistry & Photobiology, B: Biology 241 (2023) 112674

Data availability [21] Y. Zhang, Z. Yang, X. Zheng, et al., Heavy atom substituted near-infrared BODIPY
nanoparticles for photodynamic therapy, Dyes Pigments 178 (2020), 108348.
[22] N. Bai, L. Wang, Y. Xue, et al., Graded interlocks for iontronic pressure sensors with
The authors do not have permission to share data. high sensitivity and high linearity over a broad range, ACS Nano 16 (3) (2022)
4338–4347.
[23] C. Li, Y. Li, Q. Wu, et al., Multifunctional BODIPY for effective inactivation of
Acknowledgements gram-positive bacteria and promotion of wound healing, Biomater. Sci. 9 (22)
(2021) 7648–7654.
The financial support from the National Natural Science Foundation [24] H. Wen, Q. Wu, C. Li, et al., 4, 4-Difluoro-4-bora-3a, 4a-diaza-s-indacene (BDPI)-
triphenylphosphine nanoparticles as a photodynamic antibacterial agent, ACS
of China (No. 51873023), and the Science and Technology Development Appl. Nano Mater. 5 (1) (2022) 1500–1507.
Plan of Changchun City (No. 21ZY38). [25] Q. Yu, X. Huang, T. Zhang, et al., Near-infrared Aza-BODIPY dyes through
molecular surgery for enhanced photothermal and photodynamic antibacterial
therapy, Chem. Res. Chin. Univ. 37 (4) (2021) 951–959.
Appendix A. Supplementary Data [26] J.L. Wang, L. Zhang, M.J. Zhao, et al., Mitochondria-targeted BODIPY
nanoparticles for enhanced photothermal and photoacoustic imaging in vivo, ACS
Appl. Bio Mater. 4 (2) (2021) 1760–1770.
Supplementary data to this article can be found online at https://doi. [27] Z. Ruan, P. Yuan, T. Li, et al., Redox-responsive prodrug-like PEGylated
org/10.1016/j.jphotobiol.2023.112674. macrophotosensitizer nanoparticles for enhanced near-infrared imaging-guided
photodynamic therapy, Eur. J. Pharm. Biopharm. 135 (2019) 25–35.
[28] C. Grazon, J. Rieger, R. Méallet-Renault, et al., Ultrabright fluorescent polymeric
References nanoparticles made from a new family of BODIPY monomers, Macromolecules 46
(13) (2013) 5167–5176.
[1] N. Huang, X. Chen, X. Zhu, et al., Ruthenium complexes/polypeptide self- [29] F.E. Alemdaroglu, S.C. Alexander, D. Ji, et al., Poly (BODIPY) s: a new class of
assembled nanoparticles for identification of bacterial infection and targeted tunable polymeric dyes, Macromolecules 42 (17) (2009) 6529–6536.
antibacterial research, Biomaterials 141 (2017) 296–313. [30] R. Sauer, A. Turshatov, S. Baluschev, et al., One-pot production of fluorescent
[2] Y. Wang, Y. Yang, Y. Shi, et al., Antibiotic-free antibacterial strategies enabled by surface-labeled polymeric nanoparticles via miniemulsion polymerization with
nanomaterials: progress and perspectives, Adv. Mater. 32 (18) (2020) 1904106. bodipy surfmers, Macromolecules 45 (9) (2012) 3787–3796.
[3] P. Li, S. Liu, X. Yang, et al., Low-drug resistance carbon quantum dots decorated [31] S. Zhu, J. Zhang, J. Janjanam, et al., Highly water-soluble, near-infrared emissive
injectable self-healing hydrogel with potent antibiofilm property and cutaneous BODIPY polymeric dye bearing RGD peptide residues for cancer imaging, Anal.
wound healing, Chem. Eng. J. 403 (2021), 126387. Chim. Acta 758 (2013) 138–144.
[4] X. Xie, C. Mao, X. Liu, et al., Tuning the bandgap of photo-sensitive polydopamine/ [32] Y. Jin, F. Ye, M. Zeigler, et al., Near-infrared fluorescent dye-doped semiconducting
Ag3PO4/graphene oxide coating for rapid, noninvasive disinfection of implants, polymer dots, ACS Nano 5 (2) (2011) 1468–1475.
ACS Central Sci. 4 (6) (2018) 724–738. [33] T. Sun, W. Lin, W. Zhang, et al., Self-assembly of amphiphilic drug-dye conjugates
[5] C. Zhou, Q. Wang, J. Jiang, et al., Nanozybiotics: nanozyme-based antibacterials into nanoparticles for imaging and chemotherapy, Chem. An Asian J. 11 (22)
against bacterial resistance, Antibiotics 11 (3) (2022) 390. (2016) 3174–3177.
[6] R. Su, H. Yan, X. Jiang, et al., Orange-red to NIR emissive carbon dots for [34] T. Zhang, C. Ma, T. Sun, et al., Unadulterated BODIPY nanoparticles for biomedical
antimicrobial, bioimaging and bacteria diagnosis, J. Mater. Chem. B 10 (8) (2022) applications, Coord. Chem. Rev. 390 (2019) 76–85.
1250–1264. [35] Z. Li, M. Zheng, X. Guan, et al., Unadulterated BODIPY-dimer nanoparticles with
[7] Z. Yang, Y. Qiao, J. Li, et al., Novel type of water-soluble photosensitizer from high stability and good biocompatibility for cellular imaging, Nanoscale 6 (11)
Trichoderma reesei for photodynamic inactivation of gram-positive bacteria, (2014) 5662–5665.
Langmuir 36 (44) (2020) 13227–13235. [36] W. Lin, T. Sun, Z. Xie, et al., A dual-responsive nanocapsule via disulfide-induced
[8] J. Xia, Z. Li, Z. Xie, et al., Near-infrared absorbing J-aggregates of boron self-assembly for therapeutic agent delivery, Chem. Sci. 7 (3) (2016) 1846–1852.
dipyrromethene for high efficient photothermal therapy, J. Colloid Interface Sci. [37] W. Zhang, W. Lin, Q. Pei, et al., Redox-hypersensitive organic nanoparticles for
599 (2021) 476–483. selective treatment of cancer cells, Chem. Mater. 28 (12) (2016) 4440–4446.
[9] C. Li, W. Lin, S. Liu, et al., Structural optimization of organic fluorophores for [38] W. Lin, W. Zhang, T. Sun, et al., Effect of molecular structure on stability of organic
highly efficient photothermal therapy, Mater. Chem. Front. 5 (1) (2021) 284–292. nanoparticles formed by BODIPY dimers, Langmuir 32 (37) (2016) 9575–9581.
[10] L. Qiao, J. Wang, M. Zheng, et al., BODIPY-based carbon dots as fluorescent [39] Y. Liu, N. Song, L. Chen, et al., Triple-BODIPY organic nanoparticles with
nanoprobes for sensing and imaging of extreme acidity, Anal. Methods 10 (16) particular fluorescence emission, Dyes Pigments 147 (2017) 241–245.
(2018) 1863–1869. [40] J.H. Olivier, J. Widmaier, R. Ziessel, Near-infrared fluorescent nanoparticles
[11] W. Che, G. Li, J. Zhang, et al., Exploiting aggregation induced emission and twisted formed by self-assembly of lipidic (Bodipy) dyes. Chemistry–a, Eur. J. Dermatol. 17
intramolecular charge transfer in a BODIPY dye for selective sensing of fluoride in (42) (2011) 11709–11714.
aqueous medium and living cells, J. Photochem. Photobiol. A Chem. 358 (2018) [41] N.K. Allampally, A. Florian, M.J. Mayoral, et al., H-aggregates of
274–283. Oligophenyleneethynylene (OPE)-BODIPY systems in water: guest size-dependent
[12] L. Quan, T. Sun, W. Lin, et al., BODIPY fluorescent chemosensor for Cu2+ detection encapsulation mechanism and co-aggregate morphology, Chem. Eur. J. 20 (34)
and its applications in living cells: fast response and high sensitivity, J. Fluoresc. 24 (2014) 10669–10678.
(3) (2014) 841–846. [42] G. Fan, Y.X. Lin, L. Yang, et al., Co-self-assembled nanoaggregates of BODIPY
[13] Y. Su, C. Lv, Y. Zhang, et al., Fluorescent nanoparticles with ultralow chromophore amphiphiles for dual colour imaging of live cells, Chem. Commun. 51 (62) (2015)
loading for long-term tumor-targeted imaging, Acta Biomater. 111 (2020) 12447–12450.
398–405. [43] B. Matarranz, A. Sampedro, G. Daniliuc, et al., Self-assembly of a carboxyl-
[14] Y. Liu, N. Song, L. Chen, et al., Synthesis of a near-infrared BODIPY dye for functionalized BODIPY dye via hydrogen bonding, Crystals 8 (11) (2018) 436.
bioimaging and photothermal therapy, Chem. –An Asian J. 13 (8) (2018) 989–995. [44] F. An, J. Xin, C. Deng, et al., Facile synthesis of near-infrared bodipy by donor
[15] J. Zhang, L. Wang, Z. Xie, Size-tunable and crystalline bodipy nanorods for engineering for in vivo tumor targeted dual-modal imaging, J. Mater. Chem. B 9
bioimaging, ACS Biomater. Sci. Eng. 4 (6) (2017) 1969–1975. (45) (2021) 9308–9315.
[16] T. Zhang, W. Zhang, M. Zheng, et al., Near-infrared BODIPY-paclitaxel conjugates [45] Y. Zhang, Z. Yang, X. Zheng, et al., Highly efficient near-infrared BODIPY
assembling organic nanoparticles for chemotherapy and bioimaging, J. Colloid phototherapeutic nanoparticles for cancer treatment, J. Mater. Chem. B 8 (24)
Interface Sci. 514 (2018) 584–591. (2020) 5305–5311.
[17] N. Song, L. Fu, Y. Liu, et al., Rational design of BODIPY organic nanoparticles for [46] J. Zou, P. Wang, Y. Wang, et al., Penetration depth tunable BODIPY derivatives for
enhanced photodynamic/photothermal therapy, Dyes Pigments 162 (2019) pH triggered enhanced photothermal/photodynamic synergistic therapy, Chem.
295–302. Sci. 10 (1) (2019) 268–276.
[18] Z. Wang, L. Huang, Y. Yan, et al., Elucidation of the intersystem crossing [47] J. Zong, H. Peng, X. Qing, et al., pH-responsive pluronic F127–lenvatinib-
mechanism in a helical BODIPY for low-dose photodynamic therapy, Angew. encapsulated halogenated boron-dipyrromethene nanoparticles for combined
Chem. 132 (37) (2020) 16248–16255. photodynamic therapy and chemotherapy of liver cancer, ACS Omega 6 (18)
[19] N. Song, Y. Li, L. Chen, et al., BODIPY derivatives as light-induced free radical (2021) 12331–12342.
generators for hypoxic cancer treatment, J. Mater. Chem. B 7 (25) (2019) [48] W. Lin, W. Zhang, S. Liu, et al., Engineering pH-responsive BODIPY nanoparticles
3976–3981. for tumor selective multimodal imaging and phototherapy, ACS Appl. Mater.
[20] P. Gao, S. Liu, Y. Su, et al., Fluorine-doped carbon dots with intrinsic nucleus- Interfaces 11 (47) (2019) 43928–43935.
targeting ability for drug and dye delivery, Bioconjug. Chem. 31 (3) (2019) [49] Q. Shi, C. Mou, Z. Xie, et al., Exploring BODIPY derivatives as photosensitizers for
646–655. antibacterial photodynamic therapy, Photodiagn. Photodyn. Ther. 102901 (2022).

You might also like