959
views
1
recommends
+1 Recommend
1 collections
    2
    shares

      Interested in becoming an AMM published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://amm-journal.org/index.php/2023/04/26/acta-materia-medica-call-for-papers-2/

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Discovery of a pyridophenoselenazinium-based photosensitizer with high photodynamic efficacy against breast cancer cells

      research-article
      Bookmark

            Abstract

            Development of efficient photosensitizers with minimal side effects is highly desirable for photodynamic therapy. Reported herein is the discovery of a novel pyridophenoselenazinium-based NIR-I photosensitizer, termed PPSe, that efficiently generated both type I and II reactive oxygen species (ROS) upon appropriate light irradiation. PPSe exhibited potent phototoxicity as well as excellent phototherapeutic indices against several breast cancer cell lines. PPSe induced DNA damage and breast cancer cell apoptosis via photo-triggered intracellular ROS generation.

            Main article text

            1. INTRODUCTION

            Breast cancer (BC) is the most common cancer among women, and one of the leading causes of female morbidity and mortality, with 2.26 million new cases and 680,000 deaths in 2020 worldwide [13]. Thus, breast cancer remains a major public health burden. Current breast cancer therapies mainly rely on surgery, radiotherapy, chemotherapy, and targeted therapy; however, these strategies are limited by physical pain, fatal side effects, and drug resistance [46]. The situation for aggressive triple negative breast cancer (TNBC) is even worse due to its lack of progesterone receptors (PRs), human epidermal growth factor receptor 2 (HER2), and estrogen receptors (ERs) [7, 8]. Therefore, it is of great importance to develop more efficient and less toxic therapies for breast cancer treatment, especially TNBC.

            In recent years, photodynamic therapy (PDT) has attracted much interest for breast cancer treatment owing to its low systemic toxicity, minimal invasiveness, negligible drug resistance, and cost effectiveness [911]. Typically, PDT utilizes a combination of photosensitizer, light, and oxygen to destroy cancer cells. An ideal photosensitizer should be non-toxic, but possess significant ROS generation capacity upon light irradiation [12]. Photosensitizers are mainly divided into type I and II based on the mechanism of ROS generation mechanism. Upon photoirradiation, type I photosensitizers produce radical reactive oxygen species, such as superoxide anion radical (O2 •−) and hydroxyl radical (OH), via an electron transfer process, while type II photosensitizers convert non-toxic molecular oxygen into highly toxic singlet oxygen (1O2) via an energy transfer photoreaction [13, 14]. Type I photosensitizers are thought to be more efficient against solid tumors because the electron transfer pathway is less O2-dependent. A number of photosensitizers have been used in clinical applications over the past 30 years, such as porphyrin, chlorin, phthalocyanine, and indocyanine green; however, most photosensitizers used clinically are type II, which is highly O2-dependent. In addition, current photosensitizers exhibit high dark toxicity, a limited penetration depth, and are challenging to synthesize [12, 15]. Therefore, new photosensitizers with excellent efficacy and minimal side effects for photodynamic cancer therapy, especially type I photosensitizers, are still in high demand.

            Methylene blue (MB) is an FDA-approved phenothiazinium-based drug for the treatment of methemoglobinemia. Methylene blue has an excellent safety profile and has been shown to be an antitumor photosensitizer; however, the redox-active property of methylene blue results in poor intracellular ROS production capacity and limited therapeutic efficacy. In this work we report the discovery of a novel pyridophenoselenazinium-based NIR-I photosensitizer (PPSe) derived from methylene blue that exhibited potent phototoxicity along with excellent phototherapeutic indices against several breast cancer cell lines, including MDA-MB-231, MDA-MB-468, MCF-7, and 4T1 cells.

            2. MATERIALS AND METHODS

            2.1 Determination of singlet oxygen quantum yield

            Singlet oxygen production was evaluated with 1,3-diphenylisobenzofuran (DPBF). The singlet oxygen generation of PPSe was measured in ethanol solutions and MB was used as a standard. The absorbance of DPBF at 410 nm was adjusted to approximately 1.0, and the absorbance of PPSe and MB at 660 nm was adjusted between 0.2 and 0.4. Then, the absorption spectrum was measured after light irradiation (660 nm, 2 mW·cm-2) for 0, 10, 20, 30, 40, 50, 60, 70, and 80 s. DPBF solution was used as the control. The singlet oxygen quantum yield was monitored by measuring the reduction in the absorbance intensity of DPBF at 410 nm using a microplate reader (Tecan, Switzerland). The values of singlet oxygen, Φ Δ, were calculated using the following equation:

                           

            where, Φ Δ represents the singlet oxygen quantum yield of PPSe, Φ MB is the singlet oxygen quantum yield of MB (0.52 in ethanol), k represents the slope of the decrease in DPBF absorbance at 410 nm with the addition of irradiation time, and F is the correction factor (F = 1 − 10−OD). OD represents the absorbance of the solution at 660 nm.

            2.2 Superoxide anion radical (O2 •−) detection

            To determine O2 •− generation, dihydrorhodamine 123 (DHR123) was used as a superoxide anion radical probe. PPSe was added to a DHR123 aqueous solution at the same concentration (5 μM). The solutions were exposed to 660 nm irradiation (10 mW/cm2) for different times. DHR123 without PPSe was used as a control. The fluorescence intensities of solutions were measured using a spectrofluorometer and the excitation wavelength was 500 nm.

            2.3 Cell incubation

            Human breast cancer cell lines (MDA-MB-231, MCF-7, MDA-MB-468, and MDA-MB-453) and mouse breast cancer 4T1 cells were obtained from Shandong University (Jinan, China). The human breast cancer cell lines were cultured in Dulbecco’s Modified Eagle Medium (DMEM) containing 10% fetal bovine serum (FBS; Gibco, Grand Island, NY, USA) and 1% double antibiotics (penicillin/streptomycin; Gibco, Grand Island, NY, USA). And the mouse breast cancer 4T1 cell was cultured in RPMI-1640 cell medium supplemented with 10% FBS and 1% double antibiotics (penicillin/streptomycin). All the cells were incubated in a 5% CO2 humidified incubator at 37°C.

            2.4 Cellular uptake

            Cellular uptake efficiency of PPSe was determined using CytoFLEX S Flow Cytometry (Beckman Coulter, Brea, California, USA). 4T1 cells were incubated with PPSe (5 μM) for the following lengths of time: 30 min; 1 h; 3 h; 6 h; and 9 h. Then, the cells were harvested and re-suspended with phosphate buffer solution and analyzed by flow cytometry. The excitation wavelength was 638 nm and the emission collection wavelength was 712 ± 25 nm.

            2.5 Intracellular ROS generation

            The in vitro ROS generation of PPSe in 4T1 cells was assessed using 2′,7′-dichlorofluorescein diacetate (DCFH-DA) as a probe. The 4T1 cells were incubated with PPSe (0.25 μM) for 2 h at 37°C, then co-incubated with DCFH-DA (10 μM, λex = 488 nm, λem = 525 nm) for 30 min at 37 °C in the dark. 4T1 cells were washed after incubation, followed by irradiation (660 nm, 20 mW/cm2, 10 min). The cells were carefully collected and analyzed immediately using flow cytometry (Beckman). Cells in the medium without compound were used as controls, and the experiments were repeated three times.

            2.6 Cytotoxicity test

            The cytotoxicity of PPSe was carried out using the Cell Counting Kit-8 (CCK-8; Dojindo, Kumamoto, Japan) assay. MDA-MB-231, MCF-7, MDA-MB-468, MDA-MB-453, and 4T1 cells were seeded on a 96-well plate at a density of 5000 cells/well for a 24-h incubation. The photosensitizer, PPSe, was diluted into different concentrations with medium. The cells were incubated with different concentrations of PPSe for 6 h, then irradiated with a 660-nm LED lamp (20 mW/cm2, 10 min). After a 24-h incubation, the CCK-8 stop solution (10 μL) was added to each well and the cell viabilities were measured. The same concentrations of PPSe were added to cancer cells for 24 h in darkness to evaluate dark cytotoxicity. After CCK-8 treatment, the absorbance of each at 450 nm was measured using a microplate reader (Tecan) and IC50 values were calculated accordingly. The experiments were repeated three times. Cell viability rates (%) and IC50 values were analyzed using GraphPad Prism 9.0.7.

            2.7 Calcein AM/PI assay

            Calcein-AM and propidium iodide (PI) were used to study live/dead cell co-staining. 4T1 cells were seeded in 35-mm plates and cultured for 24 h. The cells were incubated with PPSe (0.25 and 0.5 μM) for 6 h, then treated with or without irradiation (660 nm, 20 mW/cm2, 10 min). Cell groups with or without irradiation were used as controls. After a 12-h incubation, the cells were first washed with phosphate buffer solution, then co-stained with calcein AM/PI for 30 min at 37 °C in the dark. Finally, all the groups of cells were imaged with a confocal laser scanning microscope (Zeiss, Germany).

            2.8 Apoptosis assays

            Cell apoptosis was analyzed using an Annexin V-FITC and PI apoptosis detection kit (Beyotime Biotechnology, Shanghai, China). 4T1 cells were cultured in 6-well Corning plates and incubated with PPSe (0.25 and 0.5 μM) at 37 °C for 6 h, then treated with or without irradiation (660 nm, 20 mW/cm2, 10 min). The 4T1 cells were incubated for 24 h after irradiation, collected, and stained with Annexin V-FITC and PI at room temperature for 30 min to analyze cell apoptosis. Cell apoptosis was detected by flow cytometry (Beckman), and the percentage of apoptotic cells was analyzed using FlowJo.

            2.9 DNA damage and apoptosis (Western blotting)

            The 4T1 cells were seeded in 6-well Corning plates at a density of 2×105 cells/well and incubated for 24 h. Then, the cells were incubated with PPSe (0.25 μM) for 6 h and irradiated with a 660-nm LED lamp (20 mW/cm2, 10 min). Cells were collected after incubation for 24 h, then lysed using RIPA lysis solution (Beyotime Biotechnology) with the addition of protease/phosphatase inhibitors for 30 min on ice. After calibrating the concentration of protein, equal amounts of protein were separated by SDS-PAGE gel electrophoresis and transferred to nitrocellulose (NC) filter membranes. The primary antibodies were mouse monoclonal caspase-3 (Proteintech, Wuhan, China) γ-H2A.X (Abcam, Cambridge, UK), and β-actin (Proteintech, Wuhan, China). Anti-mouse IgG HRP-conjugated antibody (Proteintech, Wuhan, China) was used as a secondary antibody. An ECL detection system (Tanon, Shanghai, China) was used to visualize the specific protein bands. The target protein levels were semi-quantitatively analyzed using Image J software.

            2.10 Statistical analysis

            The statistical analysis between groups was performed with one-way analysis of variance (ANOVA) and Student’s t-test using GraphPad Prism 9.0.7. software. The results are expressed as the mean ± standard deviation (SD). A p value < 0.05 was considered statistically significant (* p < 0.05, ** p < 0.01, *** p < 0.001).

            3. RESULTS AND DISCUSSION

            The photosensitizer, PPSe, was synthesized through redox-neutral annulation between readily accessible 3,3’-diselanediylbis (N, N-dimethyl-4-nitrosoaniline) and 4-[(quinolin-8-ylamino) methyl] benzoic acid, as shown in Scheme S1 .

            The optical properties of PPSe were first analyzed using UV-vis absorption and fluorescence spectra. As shown in Figure 1b , the maximum absorption of PPSe centered at approximately 660 nm. Upon 660 nm light excitation, PPSe exhibited NIR-I emission with a maximum peak at approximately 690 nm.

            Figure 1 |

            (a) The structure of the photosensitizer, PPSe. (b) UV-vis and fluorescence spectra of PPSe in ethanol. (c-d) Absorption changes of the DPBF/PPSe under 660-nm LED light irradiation (2 mW/cm2) in ethanol solution. (e) O2 •− detection using the DHR123 (5 μM) assay for PPSe (5 μM) in aqueous solution upon irradiation (660 nm, 10 mW/cm2). (f) Fluorescence response of DHR123 for PPSe (5 μM) and DHR123 (5 μM) in aqueous solution.

            The ROS generation capacity of PPSe was then evaluated because the ROS generation capacity represents the photodynamic efficacy of a photosensitizer. To determine whether PPSe generated 1O2, DPBF was used as the 1O2 capture probe and MB (Φ Δ = 0.52 in ethanol) was used as a reference ( Figures 1c, d and S1 ). As the irradiation time increased, the absorbance of the DPBF/PPSe ethanol solution decreased under 660 nm light irradiation (2 mW/cm2). The singlet oxygen quantum yield of PPSe in ethanol was determined to be 0.62, indicating that PPSe had strong singlet oxygen generation efficiency. To characterize the superoxide anion radical generation ability of PPSe in the aqueous solution upon irradiation, a DHR123 probe was used as the superoxide anion radical (O2 •−) indicator. There was no obvious fluorescence change in the DHR123 alone group under irradiation (660 nm, 10 mW/cm2), but significant fluorescence enhancement of the PPSe (5 μM) and DHR123 (5 μM) group was observed, indicating O2 •− production ( Figure 1e, 1f ). Taken together, PPSe efficiently produced ROS through electron and energy transfer processes. Thus, PPSe is a type I/II combined photosensitizer.

            To investigate the cellular uptake efficiency of PPSe, flow cytometry was used to monitor the changes in fluorescence intensity among living cells. The 4T1 cells were treated with PPSe (5 μM) at the following times: 30 min; 1 h; 3 h; 6 h; and 9 h. As shown in Figure 2a and 2b , a right shift of the chromatogram represents increased fluorescent intensity, which can indicate increased cellular uptake. It was observed that the cellular uptake of PPSe displayed a gradual increase over time, with saturation at approximately 6 h. Overall, the cellular uptake experiment showed that PPSe was efficiently taken up by breast cancer cells.

            Figure 2 |

            (a-b) Flow cytometry analysis and the curve of median fluorescence intensity of cellular uptake of PPSe (5 μM) after incubation for different time periods. (c-d) Flow cytometry analysis and the curve of median fluorescence intensity of ROS generation in 4T1 cells treated with PPSe (0.25 μM) with or without light irradiation (660 nm, 12 J/cm2). Results are expressed as mean ± SD (n=3), ***P < 0.001 vs the PBS plus light group.

            Because PPSe exhibited PDT potential by generating 1O2 and O2 •− as discussed above, the intracellular ROS produced by PPSe in living cells were further measured using DCFH-DA as a fluorescent probe. The breast cancer 4T1 cells were treated with PPSe (0.25 μM) for 6 h, followed by incubation with the DCFH-DA (10 μM) probe for 30 min. As shown in Figure 2c and 2d , there were no apparent differences in the PBS groups with or without irradiation. The intracellular ROS level in the PPSe-treated cells was negligible in the dark, but significantly increased upon 660 nm irradiation (20 mW/cm2, 10 min). The results indicated that PPSe efficiently induced ROS generation under light irradiation and might have potent phototoxicity.

            To assess the photodynamic effect of PPSe against breast cancer cells, the cytotoxicity of PPSe against 4T1 cells was investigated using the CCK-8 assay. As depicted in Figure 3a , the cell viabilities of 4T1 cells incubated with different concentrations of PPSe and exposed to different light doses (660 nm, 4 J/cm2, 8 J/cm2, 12 J/cm2) were evaluated. The results indicated that the phototoxicity of PPSe increased in a dose-dependent manner within the investigated light dose ranges, and PPSe exhibited the best photodynamic efficacy under the irradiation of 12 J/cm2 (20 mW/cm2, 10 min). Notably, PPSe exhibited excellent cytotoxicity against 4T1 cell lines with an IC50 value of 170.2 ± 9.6 nM upon 660 nm light irradiation (12 J/cm2), as shown in Figure 3b . Furthermore, the photodynamic effect of PPSe under hypoxia was also investigated. Because PPSe mainly generated 1O2 following a type II pathway, PPSe exhibited poor phototherapeutic efficacy without a dose-dependent fashion under 1% O2 conditions ( Figure S5 ). It is noteworthy that PPSe exhibited no significant dark toxicity within the tested concentrations, which indicated that PPSe has an excellent phototherapeutic index (> 58) against 4T1 cells.

            Figure 3 |

            (a) Cell viabilities of 4T1 cells incubated with various concentrations of PPSe after exposure to different light doses (660 nm). (b) Cell viabilities of 4T1 cells incubated with various concentrations of PPSe with or without irradiation (660 nm, 12 J/cm2). (c) Fluorescence images of Calcein-AM and PI co-stained 4T1 cells treated with PPSe with or without irradiation, the irradiation is 660 nm, 12 J/cm2. Scale bar: 100 μm.

            To visually confirm the cytotoxicity of PPSe, the photodynamic effect of PPSe toward cancer cells was further evaluated by the live/dead cell co-staining assay using PI and calcein-AM. Calcein-AM stains live cells to emit green fluorescence, whereas PI stains dead cells to emit red fluorescence. As shown in Figure 3c , almost no cells were dead in the PBS control groups or the groups treated with PPSe (0.25 and 0.5 μM) without light irradiation, in which a large percentage of cells were stained green by calcein-AM. By contrast, the cancer cells treated with PPSe were significantly killed in a concentration-dependent manner upon light irradiation. As shown in Figure 3c , nearly one-half of the 4T1 cells were killed when incubated with 0.25 μM PPSe plus 660 nm light irradiation (20 mW/cm2, 10 min). In addition, significant red fluorescence was observed after treating 4T1 cells with 0.5 μM PPSe plus light irradiation, indicating that nearly all the cells were efficiently killed, which correlated well with the cell viability assay findings.

            To further evaluate the cytotoxicity of PPSe against human breast cancer cells, the viability of MDA-MB-231, MCF-7, MDA-MB-468, and MDA-MB-453 cells was also determined using the CCK-8 assay. As shown in Figure 4 and Table S1 , there were slight differences among the four breast cancer cell lines. The IC50 values of PPSe against the MDA-MB-231, MCF-7, MDA-MB-468, and MDA-MB-453 human breast cancer cell lines under 660-nm light irradiation (20 mW/cm2, 10 min) conditions were 0.16 ± 0.010, 0.71 ± 0.061, 0.12 ± 0.014, and 0.29 ± 0.012 μM, respectively, which were similar to the 4T1 cell results. The dark toxicity of PPSe was also determined under the same experimental conditions except for light irradiation. It was shown that PPSe exhibited weak dark toxicity within the tested concentrations against the breast cancer cell lines. It should be noted that the phototherapeutic index of PPSe against MDA-MB-231 cells was excellent (>125).

            Figure 4 |

            MDA-MB-231 (a) MCF-7 (b) MDA-MB-468 (c) and MDA-MB-453 (d) cell viabilities after incubation with various concentrations of PPSe with or without irradiation (660 nm, 12 J/cm2).

            The flow cytometry assays with Annexin V-FITC/PI staining were performed to evaluate the apoptotic effects of PPSe on 4T1 cells. As illustrated in Figure 5a and 5b , the percentage of apoptotic cells was significantly increased in the PPSe plus irradiation (660 nm, 20 mW/cm2, 10 min) groups compared with the PPSe alone groups. The cell apoptotic rates in the PPSe (0.25 μM) alone group and the PPSe (0.25 μM) plus irradiation group were 2.31% and 17.32%, respectively. When the 4T1 cells were incubated with 0.5 μM PPSe plus irradiation, the cell apoptotic rate was significantly increased to 32.51%. By contrast, there was no significant change in the cell apoptosis rate of the PBS groups whether irradiated or not. Taken together, PPSe efficiently induced 4T1 cell apoptosis under light irradiation and the percentage of apoptotic cells increased as the concentration of PPSe increased, which is consistent with the cytotoxicity of PPSe.

            Figure 5 |

            (a-b) Effects of PPSe on cell apoptosis in 4T1 cells. Results are expressed as mean ± SD (n=3), **P < 0.01 and ***P < 0.001 all vs the PBS plus light group. (c) Western blot assay of DNA damage-relevant protein γ-H2A.X and apoptosis-relevant protein cleaved caspase-3 protein in 4T1 cells, β-actin utilized as an internal loading control. (d-e) Quantitative protein expression of cleaved caspase-3 and γ-H2A.X. All results are expressed as mean ± SD (n=3), ***P < 0.001 vs the PBS plus light group.

            To further investigate the mechanism of cell death induced by PPSe under light irradiation, we performed Western blot assays to monitor DNA damage and apoptosis in 4T1 cells. As shown in Figure 5c , the expression of cleaved-caspase-3 and γ-H2A.X increased significantly in the PPSe (0.5 μM) plus light irradiation (660 nm, 20 mW/cm2, 10 min) group, but not in the PPSe alone (0.5 μM) and PBS control groups. These results indicated that the combination of PPSe and light irradiation led to efficient DNA damage and induced apoptosis.

            4. CONCLUSIONS

            In summary, we have described the discovery and characterization of a new NIR-I photosensitizer (PPSe) that efficiently generated ROS via a type I and II combined pathway in the presence of light. PPSe exhibited potent phototoxicity against five breast cancer cell lines. It is noteworthy that the phototherapeutic index of PPSe against MDA-MB-231 cells was excellent (>125). Cell death mechanism studies indicated that PPSe induced DNA damage and cell apoptosis in the presence of light. We hope that PPSe will be considered suitable for more applications in PDT.

            Supplementary Material

            Supplementary Material can be downloaded here

            CONFLICTS OF INTEREST

            The authors declare no competing financial interest.

            REFERENCES

            1. Becker S. A Historic and Scientific Review of Breast Cancer: The Next Global Healthcare Challenge. International Journal of Gynaecology and Obstetrics: the Official Organ of the International Federation of Gynaecology and Obstetrics. 2015. Vol. 131 Suppl 1:S36–S39

            2. Henley SJ, Thomas CC, Lewis DR, Ward EM, Islami F, Wu M, et al.. Annual Report to the Nation on the Status of Cancer, Part II: Progress Toward Healthy People 2020 Objectives for 4 Common Cancers. Cancer. 2020. Vol. 126:2250–2266

            3. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al.. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians. 2021. Vol. 71:209–249

            4. Choi J, Jung WH, Koo JS. Clinicopathologic Features of Molecular Subtypes of Triple Negative Breast Cancer Based on Immunohistochemical Markers. Histology and Histopathology. 2012. Vol. 27:1481–1493

            5. Kim C, Gao R, Sei E, Brandt R, Hartman J, Hatschek T, et al.. Chemoresistance Evolution in Triple-Negative Breast Cancer Delineated by Single-Cell Sequencing. Cell. 2018. Vol. 173:879–893 e13

            6. Parise CA, Caggiano V. Breast Cancer Survival Defined by the ER/PR/HER2 Subtypes and a Surrogate Classification according to Tumor Grade and Immunohistochemical Biomarkers. Journal of Cancer Epidemiology. 2014. Vol. 2014:469251

            7. Geyer FC, Pareja F, Weigelt B, Rakha E, Ellis IO, Schnitt SJ, et al.. The Spectrum of Triple-Negative Breast Disease: High- and Low-Grade Lesions. The American Journal of Pathology. 2017. Vol. 187:2139–2151

            8. Perou CM. Molecular Stratification of Triple-negative Breast Cancers. Oncologist. 2011. Vol. 16 Suppl 1:61–70

            9. Czarnecka-Czapczynska M, Aebisher D, Oles P, Sosna B, Krupka-Olek M, Dynarowicz K, et al.. The Role of Photodynamic Therapy in Breast Cancer - A Review of In Vitro Research. Biomedicine & Pharmacotherapy. 2021. Vol. 144:112342

            10. Li X, Lovell JF, Yoon J, Chen X. Clinical Development and Potential of Photothermal and Photodynamic Therapies for Cancer. Nature Reviews. Clinical Oncology. 2020. Vol. 17:657–674

            11. Gao Z, Zheng S, Kamei K, Tian C. Recent Progress in Cancer Therapy Based on the Combination of Ferroptosis with Photodynamic Therapy. Acta Materia Medica. 2022. Vol. 1:411–426

            12. Pham TC, Nguyen VN, Choi Y, Lee S, Yoon J. Recent Strategies to Develop Innovative Photosensitizers for Enhanced Photodynamic Therapy. Chemical Reviews. 2021. Vol. 121:13454–13619

            13. Chen D, Xu Q, Wang W, Shao J, Huang W, Dong X. Type I Photosensitizers Revitalizing Photodynamic Oncotherapy. Small. 2021. Vol. 17:e2006742

            14. Wan Y, Fu LH, Li C, Lin J, Huang P. Conquering the Hypoxia Limitation for Photodynamic Therapy. Advanced Materials (Deerfield Beach, Fla.). 2021. Vol. 33:e2103978

            15. Lovell JF, Liu TW, Chen J, Zheng G. Activatable Photosensitizers for Imaging and Therapy. Chemical Reviews. 2010. Vol. 110:2839–2857

            Graphic abstract

            Highlights
            • A novel pyridophenoselenazinium-based NIR-I photosensitizer (PPSe) was developed.

            • PPSe efficiently generated both type I and II ROS upon appropriate light irradiation and exhibited high photodynamic efficacy against breast cancer cells.

            • PPSe induced DNA damage and breast cancer cell apoptosis.

            In brief

            A novel pyridophenoselenazinium-based NIR-I photosensitizer (PPSe) with potent phototoxicity as well as excellent phototherapeutic indices against breast cancer cells was developed.

            Author and article information

            Journal
            amm
            Acta Materia Medica
            Compuscript (Ireland )
            2737-7946
            23 March 2023
            : 2
            : 1
            : 96-105
            Affiliations
            [a ]Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China
            [b ]Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, P. R. China
            Author notes
            *Correspondence: gangshan@ 123456sdu.edu.cn (G. Shan); lidonghai@ 123456sdu.edu.cn (D. Li)

            #These authors contributed equally to this work.

            Article
            10.15212/AMM-2023-0002
            bbd53fb8-9596-45e1-926e-e05bcbbf8f72
            Copyright © 2023 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 13 January 2023
            : 23 February 2023
            : 02 March 2023
            Page count
            Figures: 5, References: 15, Pages: 10
            Funding
            Funded by: National Natural Science Foundation of China
            Award ID: 82003589
            Funded by: Taishan Scholars Program at Shandong Province
            Award ID: tsqn 201909036
            Funded by: Natural Science Foundation of Shandong Province
            Award ID: ZR2022QC058
            Funded by: Guangdong Basic and Applied Basic Research Foundation
            Award ID: 2022A1515110292
            The authors acknowledge the National Natural Science Foundation of China (No. 82003589), the Taishan Scholars Program at Shandong Province (No. tsqn 201909036), and the Qilu Scholar Program at Shandong University for financial support. The authors also acknowledge the Natural Science Foundation of Shandong Province (No. ZR2022QC058) and the Guangdong Basic and Applied Basic Research Foundation (2022A1515110292).
            Categories
            Research Article

            Toxicology,Pathology,Biochemistry,Clinical chemistry,Pharmaceutical chemistry,Pharmacology & Pharmaceutical medicine
            Photodynamic therapy,Breast cancer,Pyridophenoselenazinium dye,Photosensitizer

            Comments

            Comment on this article